WO2021113685A2 - Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease - Google Patents
Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease Download PDFInfo
- Publication number
- WO2021113685A2 WO2021113685A2 PCT/US2020/063378 US2020063378W WO2021113685A2 WO 2021113685 A2 WO2021113685 A2 WO 2021113685A2 US 2020063378 W US2020063378 W US 2020063378W WO 2021113685 A2 WO2021113685 A2 WO 2021113685A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- tregs
- expanded
- cryopreserved
- population
- therapeutic population
- Prior art date
Links
- 210000003289 regulatory T cell Anatomy 0.000 title claims abstract description 1226
- 238000000034 method Methods 0.000 title claims abstract description 400
- 239000000203 mixture Substances 0.000 title claims abstract description 139
- 208000015122 neurodegenerative disease Diseases 0.000 title claims abstract description 30
- 230000004770 neurodegeneration Effects 0.000 title claims description 13
- 210000004027 cell Anatomy 0.000 claims abstract description 194
- 241000282414 Homo sapiens Species 0.000 claims abstract description 75
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 claims abstract description 70
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 37
- 238000011282 treatment Methods 0.000 claims abstract description 37
- 238000004519 manufacturing process Methods 0.000 claims abstract description 33
- 208000024827 Alzheimer disease Diseases 0.000 claims abstract description 21
- 230000001225 therapeutic effect Effects 0.000 claims description 457
- 230000014509 gene expression Effects 0.000 claims description 270
- 108090000623 proteins and genes Proteins 0.000 claims description 256
- 238000010257 thawing Methods 0.000 claims description 128
- 230000001965 increasing effect Effects 0.000 claims description 115
- 238000000684 flow cytometry Methods 0.000 claims description 101
- 210000004969 inflammatory cell Anatomy 0.000 claims description 93
- 239000001963 growth medium Substances 0.000 claims description 84
- 230000006870 function Effects 0.000 claims description 74
- 239000008194 pharmaceutical composition Substances 0.000 claims description 67
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 claims description 61
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 claims description 61
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 claims description 61
- -1 IL l b Proteins 0.000 claims description 51
- 238000005138 cryopreservation Methods 0.000 claims description 50
- 238000012258 culturing Methods 0.000 claims description 50
- 230000000977 initiatory effect Effects 0.000 claims description 49
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 47
- 239000011324 bead Substances 0.000 claims description 43
- 230000035899 viability Effects 0.000 claims description 43
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 claims description 42
- 230000035755 proliferation Effects 0.000 claims description 38
- 230000003247 decreasing effect Effects 0.000 claims description 33
- 108010002350 Interleukin-2 Proteins 0.000 claims description 31
- 230000001629 suppression Effects 0.000 claims description 31
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 claims description 30
- 210000002540 macrophage Anatomy 0.000 claims description 30
- 108010021064 CTLA-4 Antigen Proteins 0.000 claims description 28
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 claims description 28
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 claims description 28
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 claims description 27
- 108090001005 Interleukin-6 Proteins 0.000 claims description 22
- 210000001616 monocyte Anatomy 0.000 claims description 22
- 102000004169 proteins and genes Human genes 0.000 claims description 22
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 claims description 21
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 claims description 21
- 208000035475 disorder Diseases 0.000 claims description 21
- 238000010348 incorporation Methods 0.000 claims description 21
- 229940104230 thymidine Drugs 0.000 claims description 21
- 238000010186 staining Methods 0.000 claims description 20
- 108700012920 TNF Proteins 0.000 claims description 19
- 230000000770 proinflammatory effect Effects 0.000 claims description 19
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 claims description 18
- 229960002930 sirolimus Drugs 0.000 claims description 18
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 claims description 18
- 102100033711 DNA replication licensing factor MCM7 Human genes 0.000 claims description 16
- 102100027581 Forkhead box protein P3 Human genes 0.000 claims description 16
- 101001018431 Homo sapiens DNA replication licensing factor MCM7 Proteins 0.000 claims description 16
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 claims description 16
- 210000004263 induced pluripotent stem cell Anatomy 0.000 claims description 16
- 208000023275 Autoimmune disease Diseases 0.000 claims description 15
- 230000001363 autoimmune Effects 0.000 claims description 15
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 claims description 14
- 102000008100 Human Serum Albumin Human genes 0.000 claims description 14
- 108091006905 Human Serum Albumin Proteins 0.000 claims description 14
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 claims description 14
- 230000017306 interleukin-6 production Effects 0.000 claims description 14
- 239000003937 drug carrier Substances 0.000 claims description 13
- 238000010353 genetic engineering Methods 0.000 claims description 13
- 108050006400 Cyclin Proteins 0.000 claims description 12
- 102100032857 Cyclin-dependent kinase 1 Human genes 0.000 claims description 12
- 101710106279 Cyclin-dependent kinase 1 Proteins 0.000 claims description 12
- 108090000652 Flap endonucleases Proteins 0.000 claims description 12
- 102100036691 Proliferating cell nuclear antigen Human genes 0.000 claims description 12
- 230000016396 cytokine production Effects 0.000 claims description 12
- 206010039073 rheumatoid arthritis Diseases 0.000 claims description 12
- 208000009329 Graft vs Host Disease Diseases 0.000 claims description 11
- 208000024908 graft versus host disease Diseases 0.000 claims description 11
- 201000011240 Frontotemporal dementia Diseases 0.000 claims description 10
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 claims description 10
- 208000018737 Parkinson disease Diseases 0.000 claims description 10
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 claims description 10
- 230000008859 change Effects 0.000 claims description 10
- 201000006417 multiple sclerosis Diseases 0.000 claims description 10
- 102100039650 ADP-ribosylation factor-like protein 2 Human genes 0.000 claims description 8
- 102100032533 ADP/ATP translocase 1 Human genes 0.000 claims description 8
- 108090001079 Adenine Nucleotide Translocator 1 Proteins 0.000 claims description 8
- 102100030233 Agmatinase, mitochondrial Human genes 0.000 claims description 8
- 102100030356 Arginase-2, mitochondrial Human genes 0.000 claims description 8
- 102100032412 Basigin Human genes 0.000 claims description 8
- 102100026596 Bcl-2-like protein 1 Human genes 0.000 claims description 8
- 101150008012 Bcl2l1 gene Proteins 0.000 claims description 8
- 102100023044 Cytosolic acyl coenzyme A thioester hydrolase Human genes 0.000 claims description 8
- 102100034001 DNA replication licensing factor MCM5 Human genes 0.000 claims description 8
- 102100033720 DNA replication licensing factor MCM6 Human genes 0.000 claims description 8
- 102100023933 Deoxyuridine 5'-triphosphate nucleotidohydrolase, mitochondrial Human genes 0.000 claims description 8
- 102100030595 HLA class II histocompatibility antigen gamma chain Human genes 0.000 claims description 8
- 108010067802 HLA-DR alpha-Chains Proteins 0.000 claims description 8
- 101000886101 Homo sapiens ADP-ribosylation factor-like protein 2 Proteins 0.000 claims description 8
- 101000792835 Homo sapiens Arginase-2, mitochondrial Proteins 0.000 claims description 8
- 101000798441 Homo sapiens Basigin Proteins 0.000 claims description 8
- 101000903587 Homo sapiens Cytosolic acyl coenzyme A thioester hydrolase Proteins 0.000 claims description 8
- 101000583807 Homo sapiens DNA replication licensing factor MCM2 Proteins 0.000 claims description 8
- 101001017545 Homo sapiens DNA replication licensing factor MCM5 Proteins 0.000 claims description 8
- 101001018484 Homo sapiens DNA replication licensing factor MCM6 Proteins 0.000 claims description 8
- 101000904652 Homo sapiens Deoxyuridine 5'-triphosphate nucleotidohydrolase, mitochondrial Proteins 0.000 claims description 8
- 101001082627 Homo sapiens HLA class II histocompatibility antigen gamma chain Proteins 0.000 claims description 8
- 101001021103 Homo sapiens Oxygen-dependent coproporphyrinogen-III oxidase, mitochondrial Proteins 0.000 claims description 8
- 101001130458 Homo sapiens Ras-related protein Ral-B Proteins 0.000 claims description 8
- 206010061218 Inflammation Diseases 0.000 claims description 8
- 208000036110 Neuroinflammatory disease Diseases 0.000 claims description 8
- 102100036201 Oxygen-dependent coproporphyrinogen-III oxidase, mitochondrial Human genes 0.000 claims description 8
- 102100031425 Ras-related protein Ral-B Human genes 0.000 claims description 8
- 108091006716 SLC25A4 Proteins 0.000 claims description 8
- 108010044012 STAT1 Transcription Factor Proteins 0.000 claims description 8
- 108010017324 STAT3 Transcription Factor Proteins 0.000 claims description 8
- 102100029904 Signal transducer and activator of transcription 1-alpha/beta Human genes 0.000 claims description 8
- 102100024040 Signal transducer and activator of transcription 3 Human genes 0.000 claims description 8
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 claims description 8
- 102100024591 Very long-chain specific acyl-CoA dehydrogenase, mitochondrial Human genes 0.000 claims description 8
- 108700000711 bcl-X Proteins 0.000 claims description 8
- 230000003959 neuroinflammation Effects 0.000 claims description 8
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 claims description 7
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 claims description 7
- 101000760730 Homo sapiens Medium-chain specific acyl-CoA dehydrogenase, mitochondrial Proteins 0.000 claims description 7
- 101000904499 Homo sapiens Transcription regulator protein BACH2 Proteins 0.000 claims description 7
- 101000760747 Homo sapiens Very long-chain specific acyl-CoA dehydrogenase, mitochondrial Proteins 0.000 claims description 7
- 102100024590 Medium-chain specific acyl-CoA dehydrogenase, mitochondrial Human genes 0.000 claims description 7
- 102100023998 Transcription regulator protein BACH2 Human genes 0.000 claims description 7
- 230000000779 depleting effect Effects 0.000 claims description 7
- 230000004064 dysfunction Effects 0.000 claims description 7
- 102100026105 3-ketoacyl-CoA thiolase, mitochondrial Human genes 0.000 claims description 6
- 208000011231 Crohn disease Diseases 0.000 claims description 6
- 208000035895 Guillain-Barré syndrome Diseases 0.000 claims description 6
- 102100036242 HLA class II histocompatibility antigen, DQ alpha 2 chain Human genes 0.000 claims description 6
- 108010086786 HLA-DQA1 antigen Proteins 0.000 claims description 6
- 206010019280 Heart failures Diseases 0.000 claims description 6
- 101000835276 Homo sapiens 3-ketoacyl-CoA thiolase, mitochondrial Proteins 0.000 claims description 6
- 206010048858 Ischaemic cardiomyopathy Diseases 0.000 claims description 6
- 101150084825 LGALSL gene Proteins 0.000 claims description 6
- 239000002577 cryoprotective agent Substances 0.000 claims description 6
- 210000000987 immune system Anatomy 0.000 claims description 6
- 210000000274 microglia Anatomy 0.000 claims description 6
- 210000000066 myeloid cell Anatomy 0.000 claims description 6
- 238000000575 proteomic method Methods 0.000 claims description 6
- 238000002054 transplantation Methods 0.000 claims description 6
- 201000004384 Alopecia Diseases 0.000 claims description 5
- 208000030767 Autoimmune encephalitis Diseases 0.000 claims description 5
- 206010003827 Autoimmune hepatitis Diseases 0.000 claims description 5
- 208000015943 Coeliac disease Diseases 0.000 claims description 5
- 206010009900 Colitis ulcerative Diseases 0.000 claims description 5
- 102100040505 HLA class II histocompatibility antigen, DR alpha chain Human genes 0.000 claims description 5
- 102100040485 HLA class II histocompatibility antigen, DRB1 beta chain Human genes 0.000 claims description 5
- 108010039343 HLA-DRB1 Chains Proteins 0.000 claims description 5
- 208000035186 Hemolytic Autoimmune Anemia Diseases 0.000 claims description 5
- 208000023105 Huntington disease Diseases 0.000 claims description 5
- 208000022559 Inflammatory bowel disease Diseases 0.000 claims description 5
- 206010033661 Pancytopenia Diseases 0.000 claims description 5
- 201000004681 Psoriasis Diseases 0.000 claims description 5
- 201000006704 Ulcerative Colitis Diseases 0.000 claims description 5
- 230000001154 acute effect Effects 0.000 claims description 5
- 230000000735 allogeneic effect Effects 0.000 claims description 5
- 231100000360 alopecia Toxicity 0.000 claims description 5
- 201000000448 autoimmune hemolytic anemia Diseases 0.000 claims description 5
- 208000024389 cytopenia Diseases 0.000 claims description 5
- 230000007812 deficiency Effects 0.000 claims description 5
- 230000004968 inflammatory condition Effects 0.000 claims description 5
- 208000017169 kidney disease Diseases 0.000 claims description 5
- 206010025135 lupus erythematosus Diseases 0.000 claims description 5
- 206010028417 myasthenia gravis Diseases 0.000 claims description 5
- 201000000596 systemic lupus erythematosus Diseases 0.000 claims description 5
- 206010043778 thyroiditis Diseases 0.000 claims description 5
- 102100030489 15-hydroxyprostaglandin dehydrogenase [NAD(+)] Human genes 0.000 claims description 4
- 102100029632 28S ribosomal protein S11, mitochondrial Human genes 0.000 claims description 4
- 102100030873 28S ribosomal protein S14, mitochondrial Human genes 0.000 claims description 4
- 102100030799 28S ribosomal protein S2, mitochondrial Human genes 0.000 claims description 4
- 102100029890 28S ribosomal protein S27, mitochondrial Human genes 0.000 claims description 4
- 102100029829 28S ribosomal protein S29, mitochondrial Human genes 0.000 claims description 4
- 102100024419 28S ribosomal protein S31, mitochondrial Human genes 0.000 claims description 4
- 102100024385 28S ribosomal protein S35, mitochondrial Human genes 0.000 claims description 4
- 102100027283 28S ribosomal protein S9, mitochondrial Human genes 0.000 claims description 4
- 102100021908 3-mercaptopyruvate sulfurtransferase Human genes 0.000 claims description 4
- 102100026163 39S ribosomal protein L12, mitochondrial Human genes 0.000 claims description 4
- 102100026146 39S ribosomal protein L13, mitochondrial Human genes 0.000 claims description 4
- 102100034036 39S ribosomal protein L22, mitochondrial Human genes 0.000 claims description 4
- 102100027561 39S ribosomal protein L37, mitochondrial Human genes 0.000 claims description 4
- 102100040297 39S ribosomal protein L39, mitochondrial Human genes 0.000 claims description 4
- 102100039776 39S ribosomal protein L4, mitochondrial Human genes 0.000 claims description 4
- 102100034140 39S ribosomal protein L43, mitochondrial Human genes 0.000 claims description 4
- 102100034147 39S ribosomal protein L44, mitochondrial Human genes 0.000 claims description 4
- 102100033752 39S ribosomal protein L46, mitochondrial Human genes 0.000 claims description 4
- 102100033746 39S ribosomal protein L48, mitochondrial Human genes 0.000 claims description 4
- 102100031585 ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Human genes 0.000 claims description 4
- 102100039646 ADP-ribosylation factor-like protein 3 Human genes 0.000 claims description 4
- 101800001241 Acetylglutamate kinase Proteins 0.000 claims description 4
- 102100022089 Acyl-[acyl-carrier-protein] hydrolase Human genes 0.000 claims description 4
- 102100022388 Acylglycerol kinase, mitochondrial Human genes 0.000 claims description 4
- 102100040439 Adenylate kinase 4, mitochondrial Human genes 0.000 claims description 4
- 102100028661 Amine oxidase [flavin-containing] A Human genes 0.000 claims description 4
- 102100022416 Aminoacyl tRNA synthase complex-interacting multifunctional protein 1 Human genes 0.000 claims description 4
- 102100022417 Aminoacyl tRNA synthase complex-interacting multifunctional protein 2 Human genes 0.000 claims description 4
- 102100035584 BRCA2 and CDKN1A-interacting protein Human genes 0.000 claims description 4
- 101100396232 Bombyx mori EN03 gene Proteins 0.000 claims description 4
- 101000964894 Bos taurus 14-3-3 protein zeta/delta Proteins 0.000 claims description 4
- 101150006869 CACYBP gene Proteins 0.000 claims description 4
- 102100029348 CDGSH iron-sulfur domain-containing protein 2 Human genes 0.000 claims description 4
- 102100039901 Calcyclin-binding protein Human genes 0.000 claims description 4
- 102100027943 Carnitine O-palmitoyltransferase 1, liver isoform Human genes 0.000 claims description 4
- 102100024853 Carnitine O-palmitoyltransferase 2, mitochondrial Human genes 0.000 claims description 4
- 102100032363 Choline dehydrogenase, mitochondrial Human genes 0.000 claims description 4
- 206010057645 Chronic Inflammatory Demyelinating Polyradiculoneuropathy Diseases 0.000 claims description 4
- 208000030939 Chronic inflammatory demyelinating polyneuropathy Diseases 0.000 claims description 4
- 102100023667 Coiled-coil domain-containing protein 124 Human genes 0.000 claims description 4
- 102100028901 Cullin-4B Human genes 0.000 claims description 4
- 108010024986 Cyclin-Dependent Kinase 2 Proteins 0.000 claims description 4
- 108010025454 Cyclin-Dependent Kinase 5 Proteins 0.000 claims description 4
- 108010025468 Cyclin-Dependent Kinase 6 Proteins 0.000 claims description 4
- 102100036239 Cyclin-dependent kinase 2 Human genes 0.000 claims description 4
- 102100026804 Cyclin-dependent kinase 6 Human genes 0.000 claims description 4
- 102100026805 Cyclin-dependent-like kinase 5 Human genes 0.000 claims description 4
- 102100030549 Cytochrome b5 type B Human genes 0.000 claims description 4
- 102100036035 Cytochrome c oxidase copper chaperone Human genes 0.000 claims description 4
- 102100022768 D-beta-hydroxybutyrate dehydrogenase, mitochondrial Human genes 0.000 claims description 4
- 102100029995 DNA ligase 1 Human genes 0.000 claims description 4
- 102100024829 DNA polymerase delta catalytic subunit Human genes 0.000 claims description 4
- 102100024823 DNA polymerase delta subunit 2 Human genes 0.000 claims description 4
- 102100039606 DNA replication licensing factor MCM3 Human genes 0.000 claims description 4
- 102100021389 DNA replication licensing factor MCM4 Human genes 0.000 claims description 4
- 102100032881 DNA-binding protein SATB1 Human genes 0.000 claims description 4
- 102100037840 Dehydrogenase/reductase SDR family member 2, mitochondrial Human genes 0.000 claims description 4
- 102100021073 Dynactin subunit 3 Human genes 0.000 claims description 4
- 102100024827 Dynamin-1-like protein Human genes 0.000 claims description 4
- 102100040565 Dynein light chain 1, cytoplasmic Human genes 0.000 claims description 4
- 102100021758 E3 ubiquitin-protein transferase MAEA Human genes 0.000 claims description 4
- 102100024604 Endoribonuclease LACTB2 Human genes 0.000 claims description 4
- 102100027681 Fructose-2,6-bisphosphatase TIGAR Human genes 0.000 claims description 4
- 102100033506 G-rich sequence factor 1 Human genes 0.000 claims description 4
- 102100027149 GDP-fucose protein O-fucosyltransferase 1 Human genes 0.000 claims description 4
- 102000000802 Galectin 3 Human genes 0.000 claims description 4
- 108010001517 Galectin 3 Proteins 0.000 claims description 4
- 102100039696 Glutamate-cysteine ligase catalytic subunit Human genes 0.000 claims description 4
- 102100030649 Histone H2B type 1-J Human genes 0.000 claims description 4
- 102100021467 Histone acetyltransferase type B catalytic subunit Human genes 0.000 claims description 4
- 101001126430 Homo sapiens 15-hydroxyprostaglandin dehydrogenase [NAD(+)] Proteins 0.000 claims description 4
- 101000728693 Homo sapiens 28S ribosomal protein S11, mitochondrial Proteins 0.000 claims description 4
- 101000635672 Homo sapiens 28S ribosomal protein S14, mitochondrial Proteins 0.000 claims description 4
- 101000636137 Homo sapiens 28S ribosomal protein S2, mitochondrial Proteins 0.000 claims description 4
- 101000727455 Homo sapiens 28S ribosomal protein S27, mitochondrial Proteins 0.000 claims description 4
- 101000727483 Homo sapiens 28S ribosomal protein S28, mitochondrial Proteins 0.000 claims description 4
- 101000727490 Homo sapiens 28S ribosomal protein S29, mitochondrial Proteins 0.000 claims description 4
- 101000689847 Homo sapiens 28S ribosomal protein S31, mitochondrial Proteins 0.000 claims description 4
- 101000689823 Homo sapiens 28S ribosomal protein S35, mitochondrial Proteins 0.000 claims description 4
- 101000694240 Homo sapiens 28S ribosomal protein S9, mitochondrial Proteins 0.000 claims description 4
- 101000753843 Homo sapiens 3-mercaptopyruvate sulfurtransferase Proteins 0.000 claims description 4
- 101000691538 Homo sapiens 39S ribosomal protein L12, mitochondrial Proteins 0.000 claims description 4
- 101000691550 Homo sapiens 39S ribosomal protein L13, mitochondrial Proteins 0.000 claims description 4
- 101000711436 Homo sapiens 39S ribosomal protein L22, mitochondrial Proteins 0.000 claims description 4
- 101000650303 Homo sapiens 39S ribosomal protein L37, mitochondrial Proteins 0.000 claims description 4
- 101001104233 Homo sapiens 39S ribosomal protein L39, mitochondrial Proteins 0.000 claims description 4
- 101000667416 Homo sapiens 39S ribosomal protein L4, mitochondrial Proteins 0.000 claims description 4
- 101000711507 Homo sapiens 39S ribosomal protein L43, mitochondrial Proteins 0.000 claims description 4
- 101000711597 Homo sapiens 39S ribosomal protein L44, mitochondrial Proteins 0.000 claims description 4
- 101000733892 Homo sapiens 39S ribosomal protein L46, mitochondrial Proteins 0.000 claims description 4
- 101000733907 Homo sapiens 39S ribosomal protein L48, mitochondrial Proteins 0.000 claims description 4
- 101000777636 Homo sapiens ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Proteins 0.000 claims description 4
- 101000886004 Homo sapiens ADP-ribosylation factor-like protein 3 Proteins 0.000 claims description 4
- 101000824278 Homo sapiens Acyl-[acyl-carrier-protein] hydrolase Proteins 0.000 claims description 4
- 101000614487 Homo sapiens Adenylate kinase 4, mitochondrial Proteins 0.000 claims description 4
- 101000652415 Homo sapiens Agmatinase, mitochondrial Proteins 0.000 claims description 4
- 101000694718 Homo sapiens Amine oxidase [flavin-containing] A Proteins 0.000 claims description 4
- 101000755762 Homo sapiens Aminoacyl tRNA synthase complex-interacting multifunctional protein 1 Proteins 0.000 claims description 4
- 101000755758 Homo sapiens Aminoacyl tRNA synthase complex-interacting multifunctional protein 2 Proteins 0.000 claims description 4
- 101000874304 Homo sapiens BRCA2 and CDKN1A-interacting protein Proteins 0.000 claims description 4
- 101000989662 Homo sapiens CDGSH iron-sulfur domain-containing protein 2 Proteins 0.000 claims description 4
- 101000859570 Homo sapiens Carnitine O-palmitoyltransferase 1, liver isoform Proteins 0.000 claims description 4
- 101000909313 Homo sapiens Carnitine O-palmitoyltransferase 2, mitochondrial Proteins 0.000 claims description 4
- 101000943088 Homo sapiens Choline dehydrogenase, mitochondrial Proteins 0.000 claims description 4
- 101000978248 Homo sapiens Coiled-coil domain-containing protein 124 Proteins 0.000 claims description 4
- 101000916231 Homo sapiens Cullin-4B Proteins 0.000 claims description 4
- 101000726631 Homo sapiens Cytochrome b5 type B Proteins 0.000 claims description 4
- 101000875933 Homo sapiens Cytochrome c oxidase copper chaperone Proteins 0.000 claims description 4
- 101000903373 Homo sapiens D-beta-hydroxybutyrate dehydrogenase, mitochondrial Proteins 0.000 claims description 4
- 101000863770 Homo sapiens DNA ligase 1 Proteins 0.000 claims description 4
- 101000909198 Homo sapiens DNA polymerase delta catalytic subunit Proteins 0.000 claims description 4
- 101000909189 Homo sapiens DNA polymerase delta subunit 2 Proteins 0.000 claims description 4
- 101000963174 Homo sapiens DNA replication licensing factor MCM3 Proteins 0.000 claims description 4
- 101000615280 Homo sapiens DNA replication licensing factor MCM4 Proteins 0.000 claims description 4
- 101000655234 Homo sapiens DNA-binding protein SATB1 Proteins 0.000 claims description 4
- 101000806149 Homo sapiens Dehydrogenase/reductase SDR family member 2, mitochondrial Proteins 0.000 claims description 4
- 101000844774 Homo sapiens Disks large-associated protein 3 Proteins 0.000 claims description 4
- 101001041187 Homo sapiens Dynactin subunit 3 Proteins 0.000 claims description 4
- 101000909218 Homo sapiens Dynamin-1-like protein Proteins 0.000 claims description 4
- 101000966403 Homo sapiens Dynein light chain 1, cytoplasmic Proteins 0.000 claims description 4
- 101000616009 Homo sapiens E3 ubiquitin-protein transferase MAEA Proteins 0.000 claims description 4
- 101001051467 Homo sapiens Endoribonuclease LACTB2 Proteins 0.000 claims description 4
- 101000651314 Homo sapiens Fructose-2,6-bisphosphatase TIGAR Proteins 0.000 claims description 4
- 101000870806 Homo sapiens G-rich sequence factor 1 Proteins 0.000 claims description 4
- 101001122376 Homo sapiens GDP-fucose protein O-fucosyltransferase 1 Proteins 0.000 claims description 4
- 101001034527 Homo sapiens Glutamate-cysteine ligase catalytic subunit Proteins 0.000 claims description 4
- 101001084678 Homo sapiens Histone H2B type 1-J Proteins 0.000 claims description 4
- 101000898976 Homo sapiens Histone acetyltransferase type B catalytic subunit Proteins 0.000 claims description 4
- 101001011441 Homo sapiens Interferon regulatory factor 4 Proteins 0.000 claims description 4
- 101001138081 Homo sapiens L-2-hydroxyglutarate dehydrogenase, mitochondrial Proteins 0.000 claims description 4
- 101001063370 Homo sapiens Legumain Proteins 0.000 claims description 4
- 101000966742 Homo sapiens Leucine-rich PPR motif-containing protein, mitochondrial Proteins 0.000 claims description 4
- 101000619640 Homo sapiens Leucine-rich repeats and immunoglobulin-like domains protein 1 Proteins 0.000 claims description 4
- 101000780208 Homo sapiens Long-chain-fatty-acid-CoA ligase 4 Proteins 0.000 claims description 4
- 101000780205 Homo sapiens Long-chain-fatty-acid-CoA ligase 5 Proteins 0.000 claims description 4
- 101000899339 Homo sapiens Lymphoid-specific helicase Proteins 0.000 claims description 4
- 101000588067 Homo sapiens Metaxin-1 Proteins 0.000 claims description 4
- 101000985328 Homo sapiens Methenyltetrahydrofolate cyclohydrolase Proteins 0.000 claims description 4
- 101000936433 Homo sapiens Methylglutaconyl-CoA hydratase, mitochondrial Proteins 0.000 claims description 4
- 101000581428 Homo sapiens Mini-chromosome maintenance complex-binding protein Proteins 0.000 claims description 4
- 101000637710 Homo sapiens Mitochondrial import inner membrane translocase subunit Tim13 Proteins 0.000 claims description 4
- 101000645277 Homo sapiens Mitochondrial import inner membrane translocase subunit Tim23 Proteins 0.000 claims description 4
- 101000801530 Homo sapiens Mitochondrial import receptor subunit TOM22 homolog Proteins 0.000 claims description 4
- 101000801539 Homo sapiens Mitochondrial import receptor subunit TOM34 Proteins 0.000 claims description 4
- 101000764216 Homo sapiens Mitochondrial import receptor subunit TOM40 homolog Proteins 0.000 claims description 4
- 101000577080 Homo sapiens Mitochondrial-processing peptidase subunit alpha Proteins 0.000 claims description 4
- 101000957259 Homo sapiens Mitotic spindle assembly checkpoint protein MAD2A Proteins 0.000 claims description 4
- 101000573234 Homo sapiens NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 8 Proteins 0.000 claims description 4
- 101000928259 Homo sapiens NADPH:adrenodoxin oxidoreductase, mitochondrial Proteins 0.000 claims description 4
- 101000598100 Homo sapiens Nuclear migration protein nudC Proteins 0.000 claims description 4
- 101000979629 Homo sapiens Nucleoside diphosphate kinase A Proteins 0.000 claims description 4
- 101000873418 Homo sapiens P-selectin glycoprotein ligand 1 Proteins 0.000 claims description 4
- 101001091191 Homo sapiens Peptidyl-prolyl cis-trans isomerase F, mitochondrial Proteins 0.000 claims description 4
- 101000827313 Homo sapiens Peptidyl-prolyl cis-trans isomerase FKBP3 Proteins 0.000 claims description 4
- 101000878221 Homo sapiens Peptidyl-prolyl cis-trans isomerase FKBP8 Proteins 0.000 claims description 4
- 101001084254 Homo sapiens Peptidyl-tRNA hydrolase 2, mitochondrial Proteins 0.000 claims description 4
- 101000689377 Homo sapiens Phospholipid scramblase 3 Proteins 0.000 claims description 4
- 101000741967 Homo sapiens Presequence protease, mitochondrial Proteins 0.000 claims description 4
- 101000609336 Homo sapiens Pyrroline-5-carboxylate reductase 2 Proteins 0.000 claims description 4
- 101001076721 Homo sapiens RNA-binding protein 38 Proteins 0.000 claims description 4
- 101001096365 Homo sapiens Replication factor C subunit 2 Proteins 0.000 claims description 4
- 101001096355 Homo sapiens Replication factor C subunit 3 Proteins 0.000 claims description 4
- 101000582404 Homo sapiens Replication factor C subunit 4 Proteins 0.000 claims description 4
- 101000582412 Homo sapiens Replication factor C subunit 5 Proteins 0.000 claims description 4
- 101001103771 Homo sapiens Ribonuclease H2 subunit A Proteins 0.000 claims description 4
- 101001103768 Homo sapiens Ribonuclease H2 subunit B Proteins 0.000 claims description 4
- 101000633778 Homo sapiens SLAM family member 5 Proteins 0.000 claims description 4
- 101000688665 Homo sapiens Sideroflexin-2 Proteins 0.000 claims description 4
- 101000831940 Homo sapiens Stathmin Proteins 0.000 claims description 4
- 101000697510 Homo sapiens Stathmin-2 Proteins 0.000 claims description 4
- 101000633445 Homo sapiens Structural maintenance of chromosomes protein 2 Proteins 0.000 claims description 4
- 101000934346 Homo sapiens T-cell surface antigen CD2 Proteins 0.000 claims description 4
- 101000852559 Homo sapiens Thioredoxin Proteins 0.000 claims description 4
- 101000844686 Homo sapiens Thioredoxin reductase 1, cytoplasmic Proteins 0.000 claims description 4
- 101000835093 Homo sapiens Transferrin receptor protein 1 Proteins 0.000 claims description 4
- 101000655141 Homo sapiens Transmembrane protein 14C Proteins 0.000 claims description 4
- 101000640976 Homo sapiens Tryptophan-tRNA ligase, cytoplasmic Proteins 0.000 claims description 4
- 101000838456 Homo sapiens Tubulin alpha-1B chain Proteins 0.000 claims description 4
- 101000713585 Homo sapiens Tubulin beta-4A chain Proteins 0.000 claims description 4
- 101000891612 Homo sapiens Tubulin beta-8 chain Proteins 0.000 claims description 4
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 claims description 4
- 101000611023 Homo sapiens Tumor necrosis factor receptor superfamily member 6 Proteins 0.000 claims description 4
- 101000873111 Homo sapiens Vesicle transport protein SEC20 Proteins 0.000 claims description 4
- 101001022836 Homo sapiens c-Myc-binding protein Proteins 0.000 claims description 4
- 101001059630 Homo sapiens m-AAA protease-interacting protein 1, mitochondrial Proteins 0.000 claims description 4
- 101150069138 HtrA2 gene Proteins 0.000 claims description 4
- 102100035692 Importin subunit alpha-1 Human genes 0.000 claims description 4
- 102100030126 Interferon regulatory factor 4 Human genes 0.000 claims description 4
- 102100020920 L-2-hydroxyglutarate dehydrogenase, mitochondrial Human genes 0.000 claims description 4
- 102100030985 Legumain Human genes 0.000 claims description 4
- 102100040589 Leucine-rich PPR motif-containing protein, mitochondrial Human genes 0.000 claims description 4
- 102100034319 Long-chain-fatty-acid-CoA ligase 4 Human genes 0.000 claims description 4
- 102100034318 Long-chain-fatty-acid-CoA ligase 5 Human genes 0.000 claims description 4
- 102100022539 Lymphoid-specific helicase Human genes 0.000 claims description 4
- 102100031603 Metaxin-1 Human genes 0.000 claims description 4
- 102100028687 Methenyltetrahydrofolate cyclohydrolase Human genes 0.000 claims description 4
- 206010049567 Miller Fisher syndrome Diseases 0.000 claims description 4
- 102100027372 Mini-chromosome maintenance complex-binding protein Human genes 0.000 claims description 4
- 102100031238 Mitochondrial dicarboxylate carrier Human genes 0.000 claims description 4
- 102100032125 Mitochondrial import inner membrane translocase subunit Tim13 Human genes 0.000 claims description 4
- 102100026255 Mitochondrial import inner membrane translocase subunit Tim23 Human genes 0.000 claims description 4
- 102100033590 Mitochondrial import receptor subunit TOM22 homolog Human genes 0.000 claims description 4
- 102100033583 Mitochondrial import receptor subunit TOM34 Human genes 0.000 claims description 4
- 102100026905 Mitochondrial import receptor subunit TOM40 homolog Human genes 0.000 claims description 4
- 102100040420 Mitochondrial thiamine pyrophosphate carrier Human genes 0.000 claims description 4
- 102100025321 Mitochondrial-processing peptidase subunit alpha Human genes 0.000 claims description 4
- 102100038792 Mitotic spindle assembly checkpoint protein MAD2A Human genes 0.000 claims description 4
- 102100026377 NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 8 Human genes 0.000 claims description 4
- 102100036777 NADPH:adrenodoxin oxidoreductase, mitochondrial Human genes 0.000 claims description 4
- 108010064862 Nicotinamide phosphoribosyltransferase Proteins 0.000 claims description 4
- 102000015532 Nicotinamide phosphoribosyltransferase Human genes 0.000 claims description 4
- 102100036965 Nuclear migration protein nudC Human genes 0.000 claims description 4
- 102100023252 Nucleoside diphosphate kinase A Human genes 0.000 claims description 4
- 208000003435 Optic Neuritis Diseases 0.000 claims description 4
- 101710148753 Ornithine aminotransferase Proteins 0.000 claims description 4
- 102100027177 Ornithine aminotransferase, mitochondrial Human genes 0.000 claims description 4
- 102100034925 P-selectin glycoprotein ligand 1 Human genes 0.000 claims description 4
- 102100034943 Peptidyl-prolyl cis-trans isomerase F, mitochondrial Human genes 0.000 claims description 4
- 102100023846 Peptidyl-prolyl cis-trans isomerase FKBP3 Human genes 0.000 claims description 4
- 102100036978 Peptidyl-prolyl cis-trans isomerase FKBP8 Human genes 0.000 claims description 4
- 102100030867 Peptidyl-tRNA hydrolase 2, mitochondrial Human genes 0.000 claims description 4
- 102100024496 Phospholipid scramblase 3 Human genes 0.000 claims description 4
- 102100038632 Presequence protease, mitochondrial Human genes 0.000 claims description 4
- 102100039450 Pyrroline-5-carboxylate reductase 2 Human genes 0.000 claims description 4
- 102100025859 RNA-binding protein 38 Human genes 0.000 claims description 4
- 101000727837 Rattus norvegicus Reduced folate transporter Proteins 0.000 claims description 4
- 102100037851 Replication factor C subunit 2 Human genes 0.000 claims description 4
- 102100037855 Replication factor C subunit 3 Human genes 0.000 claims description 4
- 102100030542 Replication factor C subunit 4 Human genes 0.000 claims description 4
- 102100030541 Replication factor C subunit 5 Human genes 0.000 claims description 4
- 102100039493 Ribonuclease H2 subunit A Human genes 0.000 claims description 4
- 102100039474 Ribonuclease H2 subunit B Human genes 0.000 claims description 4
- 102100029216 SLAM family member 5 Human genes 0.000 claims description 4
- 108091006410 SLC25A10 Proteins 0.000 claims description 4
- 108091006423 SLC25A19 Proteins 0.000 claims description 4
- 206010039710 Scleroderma Diseases 0.000 claims description 4
- 102100021117 Serine protease HTRA2, mitochondrial Human genes 0.000 claims description 4
- 102100024225 Sideroflexin-2 Human genes 0.000 claims description 4
- 102100024237 Stathmin Human genes 0.000 claims description 4
- 102100028051 Stathmin-2 Human genes 0.000 claims description 4
- 102100029540 Structural maintenance of chromosomes protein 2 Human genes 0.000 claims description 4
- 201000009594 Systemic Scleroderma Diseases 0.000 claims description 4
- 206010042953 Systemic sclerosis Diseases 0.000 claims description 4
- 230000005867 T cell response Effects 0.000 claims description 4
- 102100025237 T-cell surface antigen CD2 Human genes 0.000 claims description 4
- 102100036407 Thioredoxin Human genes 0.000 claims description 4
- 102100031208 Thioredoxin reductase 1, cytoplasmic Human genes 0.000 claims description 4
- 108010022173 Thiosulfate sulfurtransferase Proteins 0.000 claims description 4
- 102100030140 Thiosulfate:glutathione sulfurtransferase Human genes 0.000 claims description 4
- 102100026144 Transferrin receptor protein 1 Human genes 0.000 claims description 4
- 102100033022 Transmembrane protein 14C Human genes 0.000 claims description 4
- 208000030886 Traumatic Brain injury Diseases 0.000 claims description 4
- 102100034300 Tryptophan-tRNA ligase, cytoplasmic Human genes 0.000 claims description 4
- 102100028969 Tubulin alpha-1B chain Human genes 0.000 claims description 4
- 102100036788 Tubulin beta-4A chain Human genes 0.000 claims description 4
- 102100040237 Tubulin beta-8 chain Human genes 0.000 claims description 4
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 claims description 4
- 102100035030 Vesicle transport protein SEC20 Human genes 0.000 claims description 4
- 208000002552 acute disseminated encephalomyelitis Diseases 0.000 claims description 4
- 210000001185 bone marrow Anatomy 0.000 claims description 4
- 102100035161 c-Myc-binding protein Human genes 0.000 claims description 4
- 206010015037 epilepsy Diseases 0.000 claims description 4
- 108010011989 karyopherin alpha 2 Proteins 0.000 claims description 4
- 210000003734 kidney Anatomy 0.000 claims description 4
- 210000004185 liver Anatomy 0.000 claims description 4
- 230000004777 loss-of-function mutation Effects 0.000 claims description 4
- 102100028825 m-AAA protease-interacting protein 1, mitochondrial Human genes 0.000 claims description 4
- 210000003470 mitochondria Anatomy 0.000 claims description 4
- 230000035772 mutation Effects 0.000 claims description 4
- 208000010125 myocardial infarction Diseases 0.000 claims description 4
- 208000008795 neuromyelitis optica Diseases 0.000 claims description 4
- 208000020431 spinal cord injury Diseases 0.000 claims description 4
- 230000004083 survival effect Effects 0.000 claims description 4
- 208000009174 transverse myelitis Diseases 0.000 claims description 4
- 230000009529 traumatic brain injury Effects 0.000 claims description 4
- 201000001320 Atherosclerosis Diseases 0.000 claims description 3
- 208000032096 Encephalitic infection Diseases 0.000 claims description 3
- 101000744394 Homo sapiens Oxidized purine nucleoside triphosphate hydrolase Proteins 0.000 claims description 3
- 101000667643 Homo sapiens Required for meiotic nuclear division protein 1 homolog Proteins 0.000 claims description 3
- 201000009906 Meningitis Diseases 0.000 claims description 3
- 102100039792 Oxidized purine nucleoside triphosphate hydrolase Human genes 0.000 claims description 3
- 102100039800 Required for meiotic nuclear division protein 1 homolog Human genes 0.000 claims description 3
- 201000004982 autoimmune uveitis Diseases 0.000 claims description 3
- 201000009950 chronic meningitis Diseases 0.000 claims description 3
- 206010014599 encephalitis Diseases 0.000 claims description 3
- 230000007246 mechanism Effects 0.000 claims description 3
- 208000000288 neurosarcoidosis Diseases 0.000 claims description 3
- 208000005987 polymyositis Diseases 0.000 claims description 3
- 208000022076 postinfectious encephalitis Diseases 0.000 claims description 3
- 108091007504 ADAM10 Proteins 0.000 claims description 2
- 101100284398 Bos taurus BoLA-DQB gene Proteins 0.000 claims description 2
- 230000033616 DNA repair Effects 0.000 claims description 2
- 230000004543 DNA replication Effects 0.000 claims description 2
- 102100039673 Disintegrin and metalloproteinase domain-containing protein 10 Human genes 0.000 claims description 2
- 241000721454 Pemphigus Species 0.000 claims description 2
- 208000027093 acute inflammatory demyelinating polyradiculoneuropathy Diseases 0.000 claims description 2
- 230000022131 cell cycle Effects 0.000 claims description 2
- 230000032823 cell division Effects 0.000 claims description 2
- 230000004663 cell proliferation Effects 0.000 claims description 2
- 230000036755 cellular response Effects 0.000 claims description 2
- 208000010353 central nervous system vasculitis Diseases 0.000 claims description 2
- 230000036541 health Effects 0.000 claims description 2
- 102100026121 Flap endonuclease 1 Human genes 0.000 claims 3
- 102100025854 Acyl-coenzyme A thioesterase 1 Human genes 0.000 claims 1
- 101710175445 Acyl-coenzyme A thioesterase 1 Proteins 0.000 claims 1
- 101001017764 Homo sapiens Lipopolysaccharide-responsive and beige-like anchor protein Proteins 0.000 claims 1
- 102100033353 Lipopolysaccharide-responsive and beige-like anchor protein Human genes 0.000 claims 1
- 201000010099 disease Diseases 0.000 abstract description 16
- 230000001976 improved effect Effects 0.000 abstract description 11
- 208000024891 symptom Diseases 0.000 abstract description 9
- 208000012902 Nervous system disease Diseases 0.000 abstract description 3
- 208000025966 Neurological disease Diseases 0.000 abstract description 3
- 238000011321 prophylaxis Methods 0.000 abstract description 2
- 239000000047 product Substances 0.000 description 201
- 239000003795 chemical substances by application Substances 0.000 description 68
- 239000000523 sample Substances 0.000 description 62
- 150000007523 nucleic acids Chemical class 0.000 description 47
- 239000012472 biological sample Substances 0.000 description 39
- 108090000765 processed proteins & peptides Proteins 0.000 description 34
- 102000039446 nucleic acids Human genes 0.000 description 33
- 108020004707 nucleic acids Proteins 0.000 description 33
- 102000004196 processed proteins & peptides Human genes 0.000 description 30
- 102000000588 Interleukin-2 Human genes 0.000 description 27
- 239000000243 solution Substances 0.000 description 27
- 229920001184 polypeptide Polymers 0.000 description 26
- 238000001802 infusion Methods 0.000 description 24
- 102000040430 polynucleotide Human genes 0.000 description 23
- 108091033319 polynucleotide Proteins 0.000 description 23
- 239000002157 polynucleotide Substances 0.000 description 23
- 230000037396 body weight Effects 0.000 description 20
- 108020004414 DNA Proteins 0.000 description 18
- 230000000295 complement effect Effects 0.000 description 18
- 102000004889 Interleukin-6 Human genes 0.000 description 17
- 150000001875 compounds Chemical class 0.000 description 16
- 235000018102 proteins Nutrition 0.000 description 16
- 238000002560 therapeutic procedure Methods 0.000 description 14
- 102000004127 Cytokines Human genes 0.000 description 13
- 108090000695 Cytokines Proteins 0.000 description 13
- 108090001007 Interleukin-8 Proteins 0.000 description 13
- 238000001514 detection method Methods 0.000 description 13
- 239000003814 drug Substances 0.000 description 13
- 239000006143 cell culture medium Substances 0.000 description 12
- 230000002757 inflammatory effect Effects 0.000 description 12
- 239000011780 sodium chloride Substances 0.000 description 12
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 11
- 241001465754 Metazoa Species 0.000 description 11
- 108020004999 messenger RNA Proteins 0.000 description 11
- 230000008685 targeting Effects 0.000 description 11
- 210000004369 blood Anatomy 0.000 description 10
- 239000008280 blood Substances 0.000 description 10
- 239000002245 particle Substances 0.000 description 10
- 238000000926 separation method Methods 0.000 description 10
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 9
- 102000004150 Flap endonucleases Human genes 0.000 description 9
- 229940024606 amino acid Drugs 0.000 description 9
- 235000001014 amino acid Nutrition 0.000 description 9
- 150000001413 amino acids Chemical class 0.000 description 9
- 230000000694 effects Effects 0.000 description 9
- 239000000463 material Substances 0.000 description 9
- 239000002773 nucleotide Substances 0.000 description 9
- 125000003729 nucleotide group Chemical group 0.000 description 9
- 230000008569 process Effects 0.000 description 9
- 125000003275 alpha amino acid group Chemical group 0.000 description 8
- 238000004458 analytical method Methods 0.000 description 8
- 238000003556 assay Methods 0.000 description 8
- 208000010877 cognitive disease Diseases 0.000 description 8
- 230000000875 corresponding effect Effects 0.000 description 8
- 208000027061 mild cognitive impairment Diseases 0.000 description 8
- 239000012071 phase Substances 0.000 description 8
- 239000013612 plasmid Substances 0.000 description 8
- 241000894007 species Species 0.000 description 8
- 102000003816 Interleukin-13 Human genes 0.000 description 7
- 108090000176 Interleukin-13 Proteins 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 238000009396 hybridization Methods 0.000 description 7
- 150000003839 salts Chemical class 0.000 description 7
- 239000000126 substance Substances 0.000 description 7
- 229940124597 therapeutic agent Drugs 0.000 description 7
- 239000013598 vector Substances 0.000 description 7
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- 102000015696 Interleukins Human genes 0.000 description 6
- 108010063738 Interleukins Proteins 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 230000007717 exclusion Effects 0.000 description 6
- 238000009472 formulation Methods 0.000 description 6
- 229940124302 mTOR inhibitor Drugs 0.000 description 6
- 239000003628 mammalian target of rapamycin inhibitor Substances 0.000 description 6
- 239000003550 marker Substances 0.000 description 6
- 239000011886 peripheral blood Substances 0.000 description 6
- 210000005259 peripheral blood Anatomy 0.000 description 6
- 230000001105 regulatory effect Effects 0.000 description 6
- 230000028327 secretion Effects 0.000 description 6
- 238000013518 transcription Methods 0.000 description 6
- 230000035897 transcription Effects 0.000 description 6
- 102100022464 5'-nucleotidase Human genes 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- 102100030385 Granzyme B Human genes 0.000 description 5
- 101000678236 Homo sapiens 5'-nucleotidase Proteins 0.000 description 5
- 101001009603 Homo sapiens Granzyme B Proteins 0.000 description 5
- 125000000539 amino acid group Chemical group 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 239000003623 enhancer Substances 0.000 description 5
- 229940088598 enzyme Drugs 0.000 description 5
- 230000004957 immunoregulator effect Effects 0.000 description 5
- 229920000642 polymer Polymers 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 238000010200 validation analysis Methods 0.000 description 5
- 101150008021 80 gene Proteins 0.000 description 4
- 102000053642 Catalytic RNA Human genes 0.000 description 4
- 108090000994 Catalytic RNA Proteins 0.000 description 4
- 206010061818 Disease progression Diseases 0.000 description 4
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 4
- 101000834898 Homo sapiens Alpha-synuclein Proteins 0.000 description 4
- 101000611936 Homo sapiens Programmed cell death protein 1 Proteins 0.000 description 4
- 101000652359 Homo sapiens Spermatogenesis-associated protein 2 Proteins 0.000 description 4
- 108091028043 Nucleic acid sequence Proteins 0.000 description 4
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 4
- 108020004459 Small interfering RNA Proteins 0.000 description 4
- 102000040945 Transcription factor Human genes 0.000 description 4
- 108091023040 Transcription factor Proteins 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 239000007864 aqueous solution Substances 0.000 description 4
- 229910002092 carbon dioxide Inorganic materials 0.000 description 4
- 239000003153 chemical reaction reagent Substances 0.000 description 4
- 238000003501 co-culture Methods 0.000 description 4
- 230000001010 compromised effect Effects 0.000 description 4
- 238000010790 dilution Methods 0.000 description 4
- 239000012895 dilution Substances 0.000 description 4
- 230000005750 disease progression Effects 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- 238000011194 good manufacturing practice Methods 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 238000002955 isolation Methods 0.000 description 4
- 210000004698 lymphocyte Anatomy 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 239000002679 microRNA Substances 0.000 description 4
- 230000010076 replication Effects 0.000 description 4
- 108091092562 ribozyme Proteins 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 239000004055 small Interfering RNA Substances 0.000 description 4
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 3
- 101150044182 8 gene Proteins 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 241000283690 Bos taurus Species 0.000 description 3
- 102000053602 DNA Human genes 0.000 description 3
- 102100025413 Formyltetrahydrofolate synthetase Human genes 0.000 description 3
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 108020005187 Oligonucleotide Probes Proteins 0.000 description 3
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 239000002585 base Substances 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 229960002685 biotin Drugs 0.000 description 3
- 239000011616 biotin Substances 0.000 description 3
- 239000001569 carbon dioxide Substances 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 238000004590 computer program Methods 0.000 description 3
- 230000007423 decrease Effects 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 108010022790 formyl-methenyl-methylenetetrahydrofolate synthetase Proteins 0.000 description 3
- 239000003018 immunosuppressive agent Substances 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 244000144972 livestock Species 0.000 description 3
- 230000005291 magnetic effect Effects 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 239000011859 microparticle Substances 0.000 description 3
- 229910052757 nitrogen Inorganic materials 0.000 description 3
- 239000002751 oligonucleotide probe Substances 0.000 description 3
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 3
- 230000002285 radioactive effect Effects 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 230000000717 retained effect Effects 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 210000001519 tissue Anatomy 0.000 description 3
- 238000012546 transfer Methods 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- 101150110188 30 gene Proteins 0.000 description 2
- 101150017816 40 gene Proteins 0.000 description 2
- VDABVNMGKGUPEY-UHFFFAOYSA-N 6-carboxyfluorescein succinimidyl ester Chemical compound C=1C(O)=CC=C2C=1OC1=CC(O)=CC=C1C2(C1=C2)OC(=O)C1=CC=C2C(=O)ON1C(=O)CCC1=O VDABVNMGKGUPEY-UHFFFAOYSA-N 0.000 description 2
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 241000972773 Aulopiformes Species 0.000 description 2
- 238000010356 CRISPR-Cas9 genome editing Methods 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 241000700198 Cavia Species 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 2
- 108091026890 Coding region Proteins 0.000 description 2
- 241000699800 Cricetinae Species 0.000 description 2
- RGHNJXZEOKUKBD-SQOUGZDYSA-N D-gluconic acid Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C(O)=O RGHNJXZEOKUKBD-SQOUGZDYSA-N 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- 241000283086 Equidae Species 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 2
- 108010001498 Galectin 1 Proteins 0.000 description 2
- 102100021736 Galectin-1 Human genes 0.000 description 2
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 2
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 2
- 102000001398 Granzyme Human genes 0.000 description 2
- 108060005986 Granzyme Proteins 0.000 description 2
- 102100036241 HLA class II histocompatibility antigen, DQ beta 1 chain Human genes 0.000 description 2
- 108010065026 HLA-DQB1 antigen Proteins 0.000 description 2
- 241001272567 Hominoidea Species 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 2
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 2
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 2
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 2
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 2
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 2
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 2
- 101150097381 Mtor gene Proteins 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 241000282579 Pan Species 0.000 description 2
- 241001494479 Pecora Species 0.000 description 2
- 108091093037 Peptide nucleic acid Proteins 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 241000282405 Pongo abelii Species 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 2
- 238000011529 RT qPCR Methods 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 102100023085 Serine/threonine-protein kinase mTOR Human genes 0.000 description 2
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 2
- 102000039471 Small Nuclear RNA Human genes 0.000 description 2
- 102000042773 Small Nucleolar RNA Human genes 0.000 description 2
- 108020003224 Small Nucleolar RNA Proteins 0.000 description 2
- 108091060271 Small temporal RNA Proteins 0.000 description 2
- 241000282887 Suidae Species 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 2
- 108700029229 Transcriptional Regulatory Elements Proteins 0.000 description 2
- 108020004566 Transfer RNA Proteins 0.000 description 2
- 108700019146 Transgenes Proteins 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 230000004075 alteration Effects 0.000 description 2
- 230000001745 anti-biotin effect Effects 0.000 description 2
- 230000000692 anti-sense effect Effects 0.000 description 2
- 239000000074 antisense oligonucleotide Substances 0.000 description 2
- 238000012230 antisense oligonucleotides Methods 0.000 description 2
- 208000010668 atopic eczema Diseases 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 238000010322 bone marrow transplantation Methods 0.000 description 2
- 230000003139 buffering effect Effects 0.000 description 2
- 235000011089 carbon dioxide Nutrition 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 229940127089 cytotoxic agent Drugs 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 238000011038 discontinuous diafiltration by volume reduction Methods 0.000 description 2
- 239000002158 endotoxin Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 230000003203 everyday effect Effects 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 238000010362 genome editing Methods 0.000 description 2
- 125000003827 glycol group Chemical group 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 230000003434 inspiratory effect Effects 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 229920006008 lipopolysaccharide Polymers 0.000 description 2
- 238000002826 magnetic-activated cell sorting Methods 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 108091070501 miRNA Proteins 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 239000002105 nanoparticle Substances 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 230000005298 paramagnetic effect Effects 0.000 description 2
- 239000013610 patient sample Substances 0.000 description 2
- 230000000704 physical effect Effects 0.000 description 2
- 210000002381 plasma Anatomy 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 230000000750 progressive effect Effects 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 235000019515 salmon Nutrition 0.000 description 2
- 108091029842 small nuclear ribonucleic acid Proteins 0.000 description 2
- 239000001488 sodium phosphate Substances 0.000 description 2
- 229910000162 sodium phosphate Inorganic materials 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 230000002992 thymic effect Effects 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 230000014616 translation Effects 0.000 description 2
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 2
- 239000012808 vapor phase Substances 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- BJEPYKJPYRNKOW-REOHCLBHSA-N (S)-malic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O BJEPYKJPYRNKOW-REOHCLBHSA-N 0.000 description 1
- TUSDEZXZIZRFGC-UHFFFAOYSA-N 1-O-galloyl-3,6-(R)-HHDP-beta-D-glucose Natural products OC1C(O2)COC(=O)C3=CC(O)=C(O)C(O)=C3C3=C(O)C(O)=C(O)C=C3C(=O)OC1C(O)C2OC(=O)C1=CC(O)=C(O)C(O)=C1 TUSDEZXZIZRFGC-UHFFFAOYSA-N 0.000 description 1
- LNETULKMXZVUST-UHFFFAOYSA-N 1-naphthoic acid Chemical compound C1=CC=C2C(C(=O)O)=CC=CC2=C1 LNETULKMXZVUST-UHFFFAOYSA-N 0.000 description 1
- ASJSAQIRZKANQN-CRCLSJGQSA-N 2-deoxy-D-ribose Chemical compound OC[C@@H](O)[C@@H](O)CC=O ASJSAQIRZKANQN-CRCLSJGQSA-N 0.000 description 1
- ZOOGRGPOEVQQDX-UUOKFMHZSA-N 3',5'-cyclic GMP Chemical compound C([C@H]1O2)OP(O)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=C(NC2=O)N)=C2N=C1 ZOOGRGPOEVQQDX-UUOKFMHZSA-N 0.000 description 1
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 description 1
- 101150066375 35 gene Proteins 0.000 description 1
- 102100026433 39S ribosomal protein L14, mitochondrial Human genes 0.000 description 1
- 102100027556 39S ribosomal protein L17, mitochondrial Human genes 0.000 description 1
- 238000010600 3H thymidine incorporation assay Methods 0.000 description 1
- POQWZGBUVXRKIR-UHFFFAOYSA-N 5-[1-[3-(azacyclotridec-1-yl)propyl]-4-(3,4-dihydroxy-5-nitrophenyl)pyrrol-3-yl]-3-nitrobenzene-1,2-diol Chemical group [O-][N+](=O)C1=C(O)C(O)=CC(C=2C(=CN(CCCN3CCCCCCCCCCCC3)C=2)C=2C=C(C(O)=C(O)C=2)[N+]([O-])=O)=C1 POQWZGBUVXRKIR-UHFFFAOYSA-N 0.000 description 1
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 1
- 101150088569 85 gene Proteins 0.000 description 1
- BUROJSBIWGDYCN-GAUTUEMISA-N AP 23573 Chemical compound C1C[C@@H](OP(C)(C)=O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 BUROJSBIWGDYCN-GAUTUEMISA-N 0.000 description 1
- KVLFRAWTRWDEDF-IRXDYDNUSA-N AZD-8055 Chemical compound C1=C(CO)C(OC)=CC=C1C1=CC=C(C(=NC(=N2)N3[C@H](COCC3)C)N3[C@H](COCC3)C)C2=N1 KVLFRAWTRWDEDF-IRXDYDNUSA-N 0.000 description 1
- 208000035657 Abasia Diseases 0.000 description 1
- 206010067484 Adverse reaction Diseases 0.000 description 1
- 229920000936 Agarose Polymers 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 102100021697 Anamorsin Human genes 0.000 description 1
- 108091023037 Aptamer Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- 241000510930 Brachyspira pilosicoli Species 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- RGHNJXZEOKUKBD-UHFFFAOYSA-N D-gluconic acid Natural products OCC(O)C(O)C(O)C(O)C(O)=O RGHNJXZEOKUKBD-UHFFFAOYSA-N 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- 201000004624 Dermatitis Diseases 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- 108010016626 Dipeptides Proteins 0.000 description 1
- 206010013142 Disinhibition Diseases 0.000 description 1
- 102100029722 Ectonucleoside triphosphate diphosphohydrolase 1 Human genes 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- HKVAMNSJSFKALM-GKUWKFKPSA-N Everolimus Chemical compound C1C[C@@H](OCCO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 HKVAMNSJSFKALM-GKUWKFKPSA-N 0.000 description 1
- 239000001263 FEMA 3042 Substances 0.000 description 1
- 101150027879 FOXP3 gene Proteins 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 108091092584 GDNA Proteins 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 101000692875 Homo sapiens 39S ribosomal protein L14, mitochondrial Proteins 0.000 description 1
- 101000650291 Homo sapiens 39S ribosomal protein L17, mitochondrial Proteins 0.000 description 1
- 101000896743 Homo sapiens Anamorsin Proteins 0.000 description 1
- 101001012447 Homo sapiens Ectonucleoside triphosphate diphosphohydrolase 1 Proteins 0.000 description 1
- 101001076407 Homo sapiens Interleukin-1 receptor antagonist protein Proteins 0.000 description 1
- 101001117317 Homo sapiens Programmed cell death 1 ligand 1 Proteins 0.000 description 1
- 101001117312 Homo sapiens Programmed cell death 1 ligand 2 Proteins 0.000 description 1
- 102100026018 Interleukin-1 receptor antagonist protein Human genes 0.000 description 1
- 108090000174 Interleukin-10 Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 241000219745 Lupinus Species 0.000 description 1
- 102000008072 Lymphokines Human genes 0.000 description 1
- 108010074338 Lymphokines Proteins 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 102000029749 Microtubule Human genes 0.000 description 1
- 108091022875 Microtubule Proteins 0.000 description 1
- 102000013967 Monokines Human genes 0.000 description 1
- 108010050619 Monokines Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 229930182474 N-glycoside Natural products 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 229930012538 Paclitaxel Natural products 0.000 description 1
- 229920002230 Pectic acid Polymers 0.000 description 1
- LRBQNJMCXXYXIU-PPKXGCFTSA-N Penta-digallate-beta-D-glucose Natural products OC1=C(O)C(O)=CC(C(=O)OC=2C(=C(O)C=C(C=2)C(=O)OC[C@@H]2[C@H]([C@H](OC(=O)C=3C=C(OC(=O)C=4C=C(O)C(O)=C(O)C=4)C(O)=C(O)C=3)[C@@H](OC(=O)C=3C=C(OC(=O)C=4C=C(O)C(O)=C(O)C=4)C(O)=C(O)C=3)[C@H](OC(=O)C=3C=C(OC(=O)C=4C=C(O)C(O)=C(O)C=4)C(O)=C(O)C=3)O2)OC(=O)C=2C=C(OC(=O)C=3C=C(O)C(O)=C(O)C=3)C(O)=C(O)C=2)O)=C1 LRBQNJMCXXYXIU-PPKXGCFTSA-N 0.000 description 1
- 108010038988 Peptide Hormones Proteins 0.000 description 1
- 102000015731 Peptide Hormones Human genes 0.000 description 1
- 108010020346 Polyglutamic Acid Proteins 0.000 description 1
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 1
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- OFOBLEOULBTSOW-UHFFFAOYSA-N Propanedioic acid Natural products OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 1
- KDCGOANMDULRCW-UHFFFAOYSA-N Purine Natural products N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 1
- 238000003559 RNA-seq method Methods 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 108091027981 Response element Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- XUIMIQQOPSSXEZ-UHFFFAOYSA-N Silicon Chemical compound [Si] XUIMIQQOPSSXEZ-UHFFFAOYSA-N 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- CBPNZQVSJQDFBE-FUXHJELOSA-N Temsirolimus Chemical compound C1C[C@@H](OC(=O)C(C)(CO)CO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 CBPNZQVSJQDFBE-FUXHJELOSA-N 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 1
- 108010046334 Urease Proteins 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 206010047115 Vasculitis Diseases 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- 229940122803 Vinca alkaloid Drugs 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 235000011054 acetic acid Nutrition 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 238000011467 adoptive cell therapy Methods 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 230000006838 adverse reaction Effects 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- PYMYPHUHKUWMLA-LMVFSUKVSA-N aldehydo-D-ribose Chemical compound OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- 239000003513 alkali Substances 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-N alpha-hydroxysuccinic acid Natural products OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 229940045799 anthracyclines and related substance Drugs 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000002555 anti-neurodegenerative effect Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 238000002617 apheresis Methods 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 229910052788 barium Inorganic materials 0.000 description 1
- DSAJWYNOEDNPEQ-UHFFFAOYSA-N barium atom Chemical compound [Ba] DSAJWYNOEDNPEQ-UHFFFAOYSA-N 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 238000003766 bioinformatics method Methods 0.000 description 1
- 230000003592 biomimetic effect Effects 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 229910052797 bismuth Inorganic materials 0.000 description 1
- JCXGWMGPZLAOME-UHFFFAOYSA-N bismuth atom Chemical compound [Bi] JCXGWMGPZLAOME-UHFFFAOYSA-N 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 239000010839 body fluid Substances 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- KDYFGRWQOYBRFD-NUQCWPJISA-N butanedioic acid Chemical compound O[14C](=O)CC[14C](O)=O KDYFGRWQOYBRFD-NUQCWPJISA-N 0.000 description 1
- 229910052793 cadmium Inorganic materials 0.000 description 1
- BDOSMKKIYDKNTQ-UHFFFAOYSA-N cadmium atom Chemical compound [Cd] BDOSMKKIYDKNTQ-UHFFFAOYSA-N 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 239000002458 cell surface marker Substances 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 108091092328 cellular RNA Proteins 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 230000000973 chemotherapeutic effect Effects 0.000 description 1
- 235000013330 chicken meat Nutrition 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 238000011281 clinical therapy Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229910017052 cobalt Inorganic materials 0.000 description 1
- 239000010941 cobalt Substances 0.000 description 1
- GUTLYIVDDKVIGB-UHFFFAOYSA-N cobalt atom Chemical compound [Co] GUTLYIVDDKVIGB-UHFFFAOYSA-N 0.000 description 1
- 239000000084 colloidal system Substances 0.000 description 1
- 238000004891 communication Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 239000002254 cytotoxic agent Substances 0.000 description 1
- 231100000599 cytotoxic agent Toxicity 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- 230000002354 daily effect Effects 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 230000007850 degeneration Effects 0.000 description 1
- 230000017858 demethylation Effects 0.000 description 1
- 238000010520 demethylation reaction Methods 0.000 description 1
- 206010061811 demyelinating polyneuropathy Diseases 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 239000005549 deoxyribonucleoside Substances 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 201000001981 dermatomyositis Diseases 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 230000006334 disulfide bridging Effects 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 229960005167 everolimus Drugs 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 235000011087 fumaric acid Nutrition 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- LRBQNJMCXXYXIU-QWKBTXIPSA-N gallotannic acid Chemical compound OC1=C(O)C(O)=CC(C(=O)OC=2C(=C(O)C=C(C=2)C(=O)OC[C@H]2[C@@H]([C@@H](OC(=O)C=3C=C(OC(=O)C=4C=C(O)C(O)=C(O)C=4)C(O)=C(O)C=3)[C@H](OC(=O)C=3C=C(OC(=O)C=4C=C(O)C(O)=C(O)C=4)C(O)=C(O)C=3)[C@@H](OC(=O)C=3C=C(OC(=O)C=4C=C(O)C(O)=C(O)C=4)C(O)=C(O)C=3)O2)OC(=O)C=2C=C(OC(=O)C=3C=C(O)C(O)=C(O)C=3)C(O)=C(O)C=2)O)=C1 LRBQNJMCXXYXIU-QWKBTXIPSA-N 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000003862 glucocorticoid Substances 0.000 description 1
- 239000000174 gluconic acid Substances 0.000 description 1
- 235000012208 gluconic acid Nutrition 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 201000005787 hematologic cancer Diseases 0.000 description 1
- 208000024200 hematopoietic and lymphoid system neoplasm Diseases 0.000 description 1
- 238000011134 hematopoietic stem cell transplantation Methods 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 238000007654 immersion Methods 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 229960003444 immunosuppressant agent Drugs 0.000 description 1
- 230000001861 immunosuppressant effect Effects 0.000 description 1
- 229940125721 immunosuppressive agent Drugs 0.000 description 1
- 229940124589 immunosuppressive drug Drugs 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 208000027866 inflammatory disease Diseases 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000012417 linear regression Methods 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 239000001630 malic acid Substances 0.000 description 1
- 235000011090 malic acid Nutrition 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- YACKEPLHDIMKIO-UHFFFAOYSA-N methylphosphonic acid Chemical class CP(O)(O)=O YACKEPLHDIMKIO-UHFFFAOYSA-N 0.000 description 1
- 239000011325 microbead Substances 0.000 description 1
- 210000004688 microtubule Anatomy 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 210000002161 motor neuron Anatomy 0.000 description 1
- 230000036457 multidrug resistance Effects 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 239000002102 nanobead Substances 0.000 description 1
- YZMHQCWXYHARLS-UHFFFAOYSA-N naphthalene-1,2-disulfonic acid Chemical class C1=CC=CC2=C(S(O)(=O)=O)C(S(=O)(=O)O)=CC=C21 YZMHQCWXYHARLS-UHFFFAOYSA-N 0.000 description 1
- PSZYNBSKGUBXEH-UHFFFAOYSA-N naphthalene-1-sulfonic acid Chemical class C1=CC=C2C(S(=O)(=O)O)=CC=CC2=C1 PSZYNBSKGUBXEH-UHFFFAOYSA-N 0.000 description 1
- 230000002314 neuroinflammatory effect Effects 0.000 description 1
- 230000003962 neuroinflammatory response Effects 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 230000001293 nucleolytic effect Effects 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- 230000009437 off-target effect Effects 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000002892 organic cations Chemical class 0.000 description 1
- 235000006408 oxalic acid Nutrition 0.000 description 1
- 229960001592 paclitaxel Drugs 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 239000000813 peptide hormone Substances 0.000 description 1
- 230000003836 peripheral circulation Effects 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 102000013415 peroxidase activity proteins Human genes 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 239000010318 polygalacturonic acid Substances 0.000 description 1
- 229920002643 polyglutamic acid Polymers 0.000 description 1
- 238000006116 polymerization reaction Methods 0.000 description 1
- 210000004896 polypeptide structure Anatomy 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000001124 posttranscriptional effect Effects 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 229940002612 prodrug Drugs 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 230000006337 proteolytic cleavage Effects 0.000 description 1
- 210000001938 protoplast Anatomy 0.000 description 1
- IGFXRKMLLMBKSA-UHFFFAOYSA-N purine Chemical compound N1=C[N]C2=NC=NC2=C1 IGFXRKMLLMBKSA-UHFFFAOYSA-N 0.000 description 1
- 150000003212 purines Chemical class 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 1
- 238000005067 remediation Methods 0.000 description 1
- 239000002342 ribonucleoside Substances 0.000 description 1
- 229960001302 ridaforolimus Drugs 0.000 description 1
- 239000004017 serum-free culture medium Substances 0.000 description 1
- 229910052710 silicon Inorganic materials 0.000 description 1
- 239000010703 silicon Substances 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 230000003595 spectral effect Effects 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 238000011476 stem cell transplantation Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 210000001179 synovial fluid Anatomy 0.000 description 1
- 210000005222 synovial tissue Anatomy 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 235000015523 tannic acid Nutrition 0.000 description 1
- 229920002258 tannic acid Polymers 0.000 description 1
- 229940033123 tannic acid Drugs 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 1
- 108091035539 telomere Proteins 0.000 description 1
- 102000055501 telomere Human genes 0.000 description 1
- 210000003411 telomere Anatomy 0.000 description 1
- 229960000235 temsirolimus Drugs 0.000 description 1
- QFJCIRLUMZQUOT-UHFFFAOYSA-N temsirolimus Natural products C1CC(O)C(OC)CC1CC(C)C1OC(=O)C2CCCCN2C(=O)C(=O)C(O)(O2)C(C)CCC2CC(OC)C(C)=CC=CC=CC(C)CC(C)C(=O)C(OC)C(O)C(C)=CC(C)C(=O)C1 QFJCIRLUMZQUOT-UHFFFAOYSA-N 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- 210000001541 thymus gland Anatomy 0.000 description 1
- 230000002110 toxicologic effect Effects 0.000 description 1
- 231100000759 toxicological effect Toxicity 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 1
- 229960004528 vincristine Drugs 0.000 description 1
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 1
- 239000013603 viral vector Substances 0.000 description 1
- 238000011179 visual inspection Methods 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01N—PRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
- A01N1/00—Preservation of bodies of humans or animals, or parts thereof
- A01N1/02—Preservation of living parts
- A01N1/0205—Chemical aspects
- A01N1/021—Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
- A01N1/0221—Freeze-process protecting agents, i.e. substances protecting cells from effects of the physical process, e.g. cryoprotectants, osmolarity regulators like oncotic agents
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01N—PRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
- A01N1/00—Preservation of bodies of humans or animals, or parts thereof
- A01N1/02—Preservation of living parts
- A01N1/0278—Physical preservation processes
- A01N1/0284—Temperature processes, i.e. using a designated change in temperature over time
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/31—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/462—Cellular immunotherapy characterized by the effect or the function of the cells
- A61K39/4621—Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/46432—Nervous system antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/46433—Antigens related to auto-immune diseases; Preparations to induce self-tolerance
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
- C12N5/0637—Immunosuppressive T lymphocytes, e.g. regulatory T cells or Treg
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/04—Immunosuppressors, e.g. cyclosporin, tacrolimus
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2302—Interleukin-2 (IL-2)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/998—Proteins not provided for elsewhere
Definitions
- the present disclosure relates to the fields of medicine, molecular biology, and specifically to the manufacture of medicaments suitable for use in the treatment of mammalian neurodegenerative diseases.
- the disclosure provides improved methods for manufacturing robust, highly pure, and functional T regulatory cells useful in the treatment of diseases such as amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, and other neurological, as well as inflammatory and autoimmune diseases or dysfunctions.
- diseases such as amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, and other neurological, as well as inflammatory and autoimmune diseases or dysfunctions.
- Tregs CD4XD25 hlgh FOXP3 T regulatory cells
- Current Treg manufacturing conditions require activation and expansion protocols that are complicated and labor-intensive. Further challenges include the time required to expand the Tregs to the necessary doses and the formulation of a final cryopreserved product after expansion that maintains cellular viability, integrity and function.
- GvHD Graft- Versus-Host Disease
- autoimmune diseases including type 1 diabetes and Crohn’s disease.
- Treg therapies may be more advantageous than immunosuppressant drugs as they may limit off target effects, thus improving efficacy and minimizing adverse effects. Therefore, the development of a manufacturing process for the robust production of highly pure and functional Tregs is crucial for future applications of Treg therapies. 3. SUMMARY
- Treg adoptive cell therapies hold great promise for treating patients with a wide variety of disorders.
- a challenge to fulfilling the potential of such therapies, however, is to be able to efficiently and quickly produce large numbers of Tregs that exhibit high purity, viability and suppressive potency that can be stored and ready for administration to patients.
- the methods presented herein address this challenge, by providing methods of producing ex v/vo-expanded Treg cell populations that exhibit exemplary viability and purity and potency.
- the methods presented herein also yield cryopreserved therapeutic populations of expanded Tregs that following thawing and without further expansion maintain the desirable purity, viability and potency characteristics of the expanded Tregs prior to cryopreservation.
- the methods presented herein provide the ability to produce potent ex v/vo-expanded Treg cell populations that may be utilized as off-the-shelf therapeutics.
- the methods presented herein yield unique ex v/vo-expanded Treg cell populations.
- the Tregs described and produced herein exhibit high viability and purity as well as potent suppressive activities, characterized by both an ability to suppress T responder cells as well as a surprising ability to suppress inflammatory cell, e.g., macrophage, activity.
- the ability to inhibit inflammatory cell, e.g., macrophage, activity is absent in freshly isolated, non-expanded Tregs obtained from either healthy or disease, e.g., ALS donors.
- the ex v/vo-expanded Tregs presented herein are distinct from freshly isolated Tregs and, in fact, may be characterized as “super-supressor” Tregs. Moreover, as also demonstrated herein, the Treg cell populations described herein exhibit unique gene products signatures.
- Treg cell populations produced and described herein including, for example, treatment of neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease and frontotemporal dementia
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (1) enriching Tregs from a cell sample suspected of containing Tregs, to produce a baseline Treg cell population; (2) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (3) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs.
- the term “baseline,” or “baseline Treg cell population denotes a population of Tregs that has been enriched from a patient sample but has not yet been expanded.
- the expanded Treg cell population and the cryopreserved therapeutic population of Tregs, following thawing and without additional expansion exhibit an ability to suppress inflammatory cells, as measured by pro-inflammatory cytokine production by the inflammatory cells, wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells.
- the ability of the cryopreserved therapeutic population of Tregs, following thawing and without additional expansion, to suppress inflammatory cells is at least 70% that of the expanded Treg cell population.
- the ability to suppress inflammatory cells is measured by IL-6, TNFa, IL l b, IL8, and/or Interferon-g production by the inflammatory cells. In some embodiments, the ability to suppress inflammatory cells is measured by IL-6 production by the inflammatory cells. In some embodiments, the cryopreserved therapeutic population of Tregs, following thawing and without additional expansion, exhibits a suppressive function, wherein the suppressive function is greater than that of the baseline Treg cell population, as determined by suppression of proliferation of responder T cells.
- the suppressive function of the cryopreserved therapeutic population of Tregs, following thawing and without additional expansion is at least 25%, at least 50%, at least 75%, at least 100% at least 150%, or at least 300% greater than the suppressive function of the baseline Treg cell population as determined by suppression of proliferation of responder T cells.
- the cryopreserved therapeutic population of Tregs, following thawing and without additional expansion exhibits a suppressive function, wherein the suppressive function is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%, as determined by suppression of proliferation of responder T cells.
- the suppressive function of the cryopreserved therapeutic population of Tregs is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the suppressive function of the expanded Treg cell population before cryopreservation.
- the proliferation of responder T cells is determined by flow cytometry or thymidine incorporation.
- the viability of the cryopreserved therapeutic population of Tregs, following thawing and without additional expansion is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%, as determined by trypan blue staining.
- the viability of the cryopreserved therapeutic population of Tregs, following thawing and without additional expansion is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the viability of the expanded Treg cell population prior to the expanded Treg cell population being cryopreserved in step (c), as determined by trypan blue staining.
- the cryopreserved therapeutic population of Tregs comprises FoxP3+ Tregs wherein the proportion of FoxP3+ Tregs is increased relative to the proportion of FoxP3+ Tregs in the Tregs in the baseline Treg cell population.
- the cryopreserved therapeutic population of Tregs comprises at least 40%, at least 50%, at least 60%, at least 70%, or at least 80% or at least 90% FoxP3+ Tregs, as determined by flow cytometry.
- the cryopreserved therapeutic population of Tregs comprise FoxP3 -expressing Tregs wherein the expression of FoxP3 is increased in the Tregs relative to expression of FoxP3 in the Tregs in the baseline Treg cell population.
- the cryopreserved therapeutic population of Tregs comprises at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% CD4 + CD25 + cells, as determined by flow cytometry. In some embodiments, the cryopreserved therapeutic population of Tregs comprises fewer than 20% CD8+ cells, as determined by flow cytometry. In some embodiments, the cryopreserved therapeutic population of Tregs comprises at least 70%, at least 80%, or at least 90% CD4 + CD25 high CD127 low Tregs, as determined by flow cytometry.
- the cell sample is a leukapheresis cell sample.
- the method further comprises obtaining the cell sample from a donor by leukapheresis.
- the cell sample is not stored overnight or frozen before carrying out the enriching step (a).
- the cell sample is obtained within 30 minutes before initiation of enriching step (a).
- step (a) comprises depleting CD8+/CD19+ cells then enriching for CD25+ cells.
- step (b) is carried out within 30 minutes after step (a).
- step (b) comprises culturing the Tregs in a culture medium that comprises beads coated with anti-CD3 antibodies and anti-CD28 antibodies.
- the beads are first added to the culture medium within about 24 hours of the initiation of the culturing.
- beads coated with anti-CD3 antibodies and anti-CD28 antibodies are added to the culture medium about 14 days after beads coated with anti-CD3 antibodies and anti-CD28 antibodies were first added to the culture medium.
- step (b) further comprises adding IL-2 to the culture medium within about 6 days of the initiation of culturing. In some embodiments, step (b) further comprises replenishing the culture medium with IL-2 about every 2-3 days after IL-2 is first added to the culture medium.
- step (b) further comprises adding rapamycin to the culture medium within about 24 hours of the initiation of the culturing. In some embodiments, step (b) further comprises replenishing the culture medium with rapamycin every 2-3 days after the rapamycin is first added to the culture medium.
- the cryopreserving step (c) is carried out at least 6 days following IL-2 addition to or replenishment of the culture medium in step (b). In some embodiments, the cryopreserving step (c) is carried out about 8-25 days after the initiation of the culturing step (b).
- step(c) comprises cryopreserving the Tregs in a cryoprotectant comprising DMSO.
- the cryopreservation step (c) comprises changing the temperature of the population of Tregs in the following increments: l°C/min to 4°C, 25°C/min to -40°C, 10°C/min to -12°C, l°C/min to -40°C, and 10°C/min to - 80°C— 90°C.
- the cryopreserved therapeutic population of Tregs is frozen at a Treg density of at least 50 million cells/mL.
- the cryopreserved therapeutic population of Tregs is frozen in a total volume of 1-1.5 mL.
- the method further comprises thawing the cryopreserved therapeutic population of Tregs after cryopreservation for about 1 week, 1 month, about 3 months, about 6 months, about 9 months, about 12 months, about 18 months or about 24 months.
- the cell sample is from a human donor.
- the human donor is a healthy donor.
- the human donor is diagnosed with or suspected of having a neurodegenerative disorder.
- the neurodegenerative disorder is amyotrophic lateral sclerosis, Alzheimer’s disease, Parkinson’s disease or frontotemporal dementia.
- the population of Tregs is subjected to genetic engineering at any point of the method prior to cry opreserving step (c).
- step (b) is automated.
- step (b) takes place in a bioreactor.
- step (b) takes place in a G-REX culture system.
- the method is performed in a closed system.
- the method further comprises thawing the cryopreserved therapeutic population of Tregs and, without further expansion, placing the population into a pharmaceutical composition comprising a pharmaceutically acceptable carrier, to produce a Treg pharmaceutical composition.
- the Treg pharmaceutical composition comprises normal saline and 5% human serum albumin.
- the method further comprises administering the Treg pharmaceutical composition to a human subject.
- the Tregs in the pharmaceutical composition are autologous to the human subject.
- the human subject has been diagnosed with or is suspected of having a neurodegenerative disorder.
- the neurodegenerative disorder is amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease or frontotemporal dementia.
- provided herein is a cryopreserved therapeutic population of Tregs produced by the method provided herein.
- a pharmaceutical composition comprising a cryopreserved therapeutic population of Tregs produced by a method provided herein, following thawing and without further expansion, and a pharmaceutically acceptable carrier.
- a pharmaceutically acceptable carrier comprising a cryopreserved therapeutic population of Tregs produced by a method provided herein, following thawing and without further expansion, and a pharmaceutically acceptable carrier.
- an ex v/vo-expanded Treg cell population that exhibits an ability to suppress inflammatory cells, as measured by pro-inflammatory cytokine production by the inflammatory cells, wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells.
- the ability to suppress inflammatory cells is measured by IL-6, TNFa, IL l b, IL8, and/or Interferon-g production by the inflammatory cells.
- the ability to suppress inflammatory cells is measured by IL-6 production by the inflammatory cells.
- the Treg cell population is autologous to a human subject with ALS. In some embodiments, the Treg cell population has been expanded from a cell sample from a human subject with ALS.
- a pharmaceutical composition comprising an ex vivo- expanded Treg cell population provided herein and a pharmaceutically acceptable carrier.
- cryopreserved therapeutic population of ex v/vo-expanded Tregs that, following thawing and without additional expansion, exhibits an ability to suppress inflammatory cells, as measured by pro-inflammatory cytokine production by the inflammatory cells, wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells.
- the ability of the cryopreserved therapeutic population of ex v/vo-expanded Tregs to suppress inflammatory cells, following expansion and without additional expansion is at least 70%, that of the ex v/vo-expanded Tregs prior to cryopreservation.
- the ability to suppress inflammatory cells is measured by IL-6, TNFa, ILip, IL8, and/or Interferon-g production by the inflammatory cells. In some embodiments, the ability to suppress inflammatory cells is measured by IL-6 production by the inflammatory cells.
- the cryopreserved therapeutic population of ex vivo- expanded Tregs following thawing and without additional expansion, exhibits a suppressive function that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation.
- the cryopreserved therapeutic population of ex v/vo-expanded Tregs exhibits a suppressive function that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the suppressive function of the ex vivo- expanded Tregs prior to cryopreservation, as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation.
- the cryopreserved therapeutic population of ex vivo- expanded Tregs exhibits at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% viability, as determined by trypan blue staining.
- the cryopreserved therapeutic population of ex v/vo-expanded Tregs, following thawing and without additional expansion exhibits a viability that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the viability of the Tregs before cryopreservation, as determined by trypan blue staining.
- the cryopreserved therapeutic population of ex vivo- expanded Tregs following thawing and without additional expansion, comprises at least 40%, at least 50%, at least 60%, at least 70%, or at least 80% or at least 90% FoxP3+ Tregs, as determined by flow cytometry.
- the cryopreserved therapeutic population of ex v/vo-expanded Tregs, following thawing and without additional expansion comprises at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% CD4 + CD25 + cells, as determined by flow cytometry.
- the cryopreserved therapeutic population of ex v/vo-expanded Tregs, following thawing and without additional expansion comprises fewer than 20% CD8+ cells, as determined by flow cytometry.
- the cryopreserved therapeutic population of ex vivo- expanded Tregs, following thawing and without additional expansion comprises at least 70%, at least 80%, or at least 90% CD4 + CD25 high CD127 low Tregs, as determined by flow cytometry.
- the ex v/vo-expanded Tregs are autologous to a human subject with ALS.
- the ex v/vo-expanded Tregs have been expanded from a cell sample from a human subject with ALS.
- a pharmaceutical composition comprising the cryopreserved therapeutic population of Tregs provided herein, following thawing and without further expansion, and a pharmaceutically acceptable carrier.
- an ex v/vo-expanded Treg cell population that exhibits an ability to suppress inflammatory cells, as measured by pro-inflammatory cytokine production by the inflammatory cells, wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells, wherein the ex vivo- expanded Treg cell population has been expanded from baseline Tregs, and wherein, in the ex vivo- expanded Treg cell population: (a) expression of one or more dysfunctional baseline signature gene products listed in Table 12 and/or Table 13 is decreased relative to the expression of the one or more gene products in baseline Tregs; (b) expression of one or more dysfunctional baseline signature gene products listed in Table 14 is decreased relative to the expression of the one or more gene products in baseline Tregs; (c) expression of one or more Treg-associated signature gene products listed in Table 15 is increased relative to the expression of the one or more gene products in baseline Tregs; (d) expression of one or more mitochondria signature gene products listed in
- the ability to suppress inflammatory cells is measured by IL-6, TNFa, IL l b, IL8, and/or Interferon-g production by the inflammatory cells. In some embodiments, the ability to suppress inflammatory cells is measured by IL-6 production by the inflammatory cells.
- expression of one or more of the following gene products is increased relative to expression of the one or more gene products in baseline Tregs: ADAMIO, AIMP1, AIMP2, ARG2, BCL2L1, BSG, CD2, CD28, CD38, CD74, CD84, CTLA4, FAS, FOXP3, GCLC, HAT1, HLA-DQA1, HLA-DQB1, HLA-DRA, HLA-DRB1, HPGD, ICOS, ILIRN, IRF4, KPNA2, LGALSl, LGMN, PCNA, POFUT1, SATB1, SELPLG, STAT1, TFRC and TNFRSF18.
- expression of one or more of the following gene products is increased relative to expression of the one or more gene products in baseline Tregs: ACAA2, ACADM, ACADVL, ACOT7,ACSLl, ACSL4, ACSL5, AGK, AGMAT, AK4, ARG2, ARL2, AUH, BCL2L1, BDH1, BNIP1, CDK1, CHDH, CIAPINl, CISD2, COX17, CPOX, CPT1A, CPT2, CYB5B, DAP3, DHRS2, DNM1L, DUT, DYNLL1, ECU, FDXR, FEN1, FKBP8, GK, GRSF1, HTRA2, L2HGDH, LACTB2, LRPPRC, MAIP1, MAOA, MPST, MRPLl, MRPL12, MRPL13, MRPL14, MRPL17, MRPL22, MRPL37, MRPL
- TIMM23 TIMM23, TMEM14C, TOMM22, TOMM34, TOMM40, and TST.
- expression of one or more of the following gene products is increased relative to expression of the one or more gene products in baseline Tregs: ARL2, ARL3, BCCIP, CCDC124, CDK1, CDK2, CDK5, CDK6, CUL4B, DCTN3, FEN1, HELLS, LIG1, MAD2L1, MAEA, MCM2, MCM2, MCM3, MCM4, MCM5, MCM6, MCM7, MCMBP, NUDC, PCNA, POLD1, POLD2, RALB, RBM38, RFC2, RFC3, RFC4, RFC5, RNASEH2A, RNASEH2B, and SMC2.
- expression of one or more of the following gene products is increased relative to expression of the one or more gene products in baseline Tregs: ACAA2, ACADM, ACADVL, ACOT7, BSG, CACYBP, CD74, CDK1, CPOX, DUT, ECU, EN03, FEN1, FKBP3, HIST1H2BJ, HLA- DQA1, HLA-DRA, HLA-DRBl, LGALSl, LGALS3, MCM5, MCM6, MCM7, MTHFD1, NAMPT, NME1, NQOl, PCNA, RABIA, RALB, SLC25A4, STAT1, STMN1, STMN2, TUBA1B, TUBB4A, TUBB8, TXN, TXNRD1, and WARS.
- expression of one or more dysfunctional baseline signature gene products listed in Table 12 and/or Table 13 is decreased relative to the expression of the one or more gene products in baseline Tregs.
- an ex v/vo-expanded Treg cell population that exhibits an ability to suppress inflammatory cells, as measured by pro-inflammatory cytokine production by the inflammatory cells, wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells, wherein the ex vivo- expanded Treg cell population has been expanded from baseline Tregs, and wherein, in the ex vivo- expanded Treg cell population, expression of one or more Treg-associated signature gene products listed in Table 14 is increased relative to the expression of the one or more gene products in baseline Tregs. In some embodiments, in the ex v/vo-expanded Treg cell population expression of one or more dysfunctional baseline signature gene products listed in Table 12 and/or Table 13 is decreased relative to the expression of the one or more gene products in baseline Tregs.
- the ex v/vo-expanded Treg cell population exhibits a suppressive function, wherein the suppressive function is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation.
- the ex v/vo-expanded Treg cell population exhibits a suppressive function, wherein the suppressive function is at least 50%, at least 75%, at least 100%, or at least 150% that of baseline Tregs, as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation.
- the ex v/vo-expanded Tregs are autologous to a human subject with ALS. In some embodiments, the ex v/vo-expanded Tregs have been expanded from a cell sample from a human subject with ALS.
- the expression of the one or more gene products is changed by a log2 change of at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, or at least about 10-fold.
- expression is determined by single-shot proteomic analysis.
- a pharmaceutical composition comprising the ex v/vo-expanded Treg cell population provided herein, following thawing and without further expansion, and a pharmaceutically acceptable carrier.
- cryopreserved composition comprising a therapeutic population of ex v/vo-expanded Tregs, wherein following thawing and without further expansion, the expression of one or more of gene products listed in Table 12-Table 18 is substantially the same in therapeutic population of Tregs as the expression of the one or more gene products in the ex v/vo-expanded Tregs prior to cryopreservation.
- the one or more gene products is not also listed in Table 19.
- the one or more gene products is a gene product associated with a dysfunctional Treg phenotype, a methylation- or epigenetics-associated gene product, a mitochondria-related gene product, or a gene product associated with the cell cycle, cell division, DNA replication or DNA repair.
- the one or more gene products is known to be important for the proliferation, health, identification, and/or mechanism of Treg cells.
- the therapeutic population of ex vivo- expanded Tregs following thawing and without additional expansion, exhibits an ability to suppress inflammatory cells, as measured by pro-inflammatory cytokine production by the inflammatory cells, wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells.
- the ability of the ex v/vo-expanded Tregs to suppress inflammatory cells, following expansion and without additional expansion, is at least 70%, that of the ex v/vo-expanded Tregs prior to cryopreservation.
- the ability of the ex v/vo-expanded Tregs to suppress inflammatory cells is measured by IL-6, TNFa, IL l b, IL8, and/or Interferon-g production by the inflammatory cells.
- the ability to suppress inflammatory cells is measured by IL-6 production by the inflammatory cells.
- the therapeutic population of ex v/vo-expanded Tregs following thawing and without additional expansion, exhibits a suppressive function that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation.
- the therapeutic population of ex v/vo-expanded Tregs following thawing and without additional expansion, exhibits a suppressive function that is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the suppressive function of the ex vivo- expanded Tregs prior to cryopreservation, as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation.
- the therapeutic population of ex v/vo-expanded Tregs exhibits at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% viability, as determined by trypan blue staining.
- the therapeutic population of ex v/vo-expanded Tregs following thawing and without additional expansion, exhibits a viability that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the viability of the Tregs before cryopreservation, as determined by trypan blue staining.
- the therapeutic population of ex v/vo-expanded Tregs, following thawing and without additional expansion comprises at least 40%, at least 50%, at least 60%, at least 70%, or at least 80% or at least 90% FoxP3+ Tregs, as determined by flow cytometry.
- the therapeutic population of ex v/vo-expanded Tregs, following thawing and without additional expansion comprises at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% CD4 + CD25 + cells, as determined by flow cytometry.
- the therapeutic population of ex v/vo-expanded Tregs, following thawing and without additional expansion comprises fewer than 20% CD8+ cells, as determined by flow cytometry. In some embodiments, the therapeutic population of ex v/vo-expanded Tregs, following thawing and without additional expansion, comprises at least 70%, at least 80%, or at least 90% CD4 + CD25 high CD127 low Tregs, as determined by flow cytometry. In some embodiments, the ex v/vo-expanded Tregs are autologous to a human subject with ALS. In some embodiments, the ex v/vo-expanded Tregs have been expanded from a cell sample from a human subject with ALS. In some embodiments, gene product expression is determined by single-shot proteomic analysis.
- a pharmaceutical composition comprising the cryopreserved composition comprising a therapeutic population of ex v/vo-expanded Tregs provided herein following thawing and without further expansion, and a pharmaceutically acceptable carrier.
- a method of treating a disorder associated with Treg dysfunction comprising: administering to a subject in need of said treatment a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating a disorder associated with Treg deficiency comprising: administering to a subject in need of said treatment a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating a disorder associated with overactivation of the immune system comprising: administering to a subject in need of said treatment a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating an inflammatory condition driven by a T cell response comprising: administering to a subject in need of said treatment a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating an inflammatory condition driven by a myeloid cell response comprising: administering to a subject in need of said treatment a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the myeloid cell is a monocyte, macrophage or microglia.
- a method of treating a neurodegenerative disorder in a subject in need thereof comprising: administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the neurodegenerative disease is Amyotrophic Lateral Sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease, frontotemporal dementia or Huntington’s disease.
- a method of treating an autoimmune disorder in a subject in need thereof comprising: administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the autoimmune disorder is polymyositis, ulcerative colitis, inflammatory bowel disease, Crohn’s disease, celiac disease, systemic sclerosis (scleroderma), multiple sclerosis (MS), rheumatoid arthritis (RA), Type I diabetes, psoriasis, dermatomyosititis, systemic lupus erythematosus, cutaneous lupus, myasthenia gravis, autoimmune nephropathy, autoimmune hemolytic anemia, autoimmune cytopenia, autoimmune encephalitis, autoimmune hepatitis, autoimmune uveitis, alopecia, thyroiditis or pemphigus.
- a method of treating graft-versus-host disease in a subject in need thereof comprising: administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the subject has received a bone marrow transplant, kidney transplant or liver transplant.
- a method of improving islet graft survival in a subject in need thereof comprising: combining islet transplantation with administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating cardio-inflammation in a subject in need thereof comprising: administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the cardio-inflammation is associated with atherosclerosis, myocardial infarction, ischemic cardiomyopathy or heart failure.
- a method of treating neuroinflammation in a subject in need thereof comprising: administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the neuroinflammation is associated with stroke, acute disseminated encephalomyelitis, acute optic neuritis, acute inflammatory demyelinating polyradiculoneuropathy, chronic inflammatory demyelinating polyradiculoneuropathy, Guillain-Barre syndrome, transverse myelitis, neuromyelitis optica, epilepsy, traumatic brain injury, spinal cord injury, encephalitis, central nervous system vasculitis, neurosarcoidosis, autoimmune or post-infectious encephalitis or chronic meningitis.
- a method of treating a Tregopathy in a subject in need thereof comprising: administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the Tregopathy is caused by a FOXP3, CD25, cytotoxic T lymphocyte-associated antigen 4 (CTLA4), LPS-responsive and beige-like anchor protein (LRBA), or BTB domain and CNC homolog 2 (BACH2) gene loss-of- function mutation, or a signal transducer and activator of transcription 3 (STAT3) gain-of- function mutation.
- CTLA4 cytotoxic T lymphocyte-associated antigen 4
- LRBA LPS-responsive and beige-like anchor protein
- BACH2 BTB domain and CNC homolog 2
- the Tregs are autologous to the subject. In other embodiments of the methods of treatment provided herein, the Tregs are allogeneic to the subject.
- the composition is a pharmaceutical composition provided herein.
- Fig. 1A-1D Treg percentage and suppressive function increased during each round of Treg infusions. Arrows and vertical dotted lines represent Treg infusions. The 1 st Treg infusion was administered on week 0 and then every 2 weeks for a total of 4 infusions. The 5 th Treg infusion was administered in each participant on weeks 48, 27 and 33, respectively, and then every 4 weeks for a total of 4 infusions. (Fig. 1 A - Participant #1, Fig. IB - Participant #2 and Fig. 1C - Participant #3) The percentage of CD4 + CD25 + FOXP3 + Tregs within the total CD4 + cell population is shown.
- Treg percentages are shown at baseline (weeks -4.6, -3.0 and -4.9 in each participant, respectively), the days of the 1 st and 5 th Treg infusions, the day after each Treg infusion, every 2 weeks during each round of infusions, and 1 month after each round.
- the data point collected the day after the 4 th Treg infusion (week 6) in participant #3 was not determined due to a flow staining error.
- FIG. ID - Participant #1, Fig. IE - Participant #2 and Fig. IF - Participant #3 Treg suppressive function is shown on simultaneous days as the Treg percentage.
- Figs. 2A-2F Disease progression slowed during each round of Treg infusions and correlated with increased Treg suppressive function. Arrows and vertical dotted lines represent Treg infusions.
- Fig. 2A - Participant #1, Fig. 2B - Participant #2 and Fig. 2C - Participant #3 Clinical progression is depicted by the ALSFRS-R (white points) and AALS (black points). Clinical progression lines during each round of Treg infusions are enlarged in side panels for the early (1) and later (2) stages of disease.
- Figs. 3A-3F Maximal inspiratory pressures stabilized during Treg infusions.
- FIG. 3A - Participant #1, Fig. 3B - Participant #2 and Fig. 3C - Participant #3 The forced vital capacity (FVC) measurements are represented as % predicted values. Measurements are shown at baseline (weeks -4.6, -3.0 and -4.9 in each participant, respectively), immediately prior to each Treg infusion, every 2 weeks during each round of infusions, and 1 month after each round. A solid gray line connects the points between each round of infusions.
- FIG. 3D - Participant #1, Fig. 3E - Participant #2 and Fig. 3F - Participant #3 Measurements of maximal inspiratory pressure (MIP) are shown in cm FhO. MIPs are shown at the same time points as FVC measurements. The MIP values were erroneously not determined for Participant #1 immediately prior to the 5 th Treg infusion and for Participant #3 one month after the second round of infusions.
- MIP maximal inspiratory pressure
- FIG. 4A Gating strategy to identify CD4 and CD8 suppressor T cells; Lymphocytes were delineated by forward/side scatter gating. CD3 was used to identify T cells among the previously selected viable lymphocytes. CD4 T cells and CD8 T cells were identified as uniquely expressing CD4 or CD8 antigens. Tregs were defined as CD4 T cells co-expressing CD25 and FOXP3. The expressions of CD25 and FoxP3 were then determined on CD8 T cells.
- FIG. 4B CD4 + FOXP3 + CD25 high T cell percentage (% of total CD4), did not differ among HC, MCI and Alzheimer groups.
- FIG. 4A Gating strategy to identify CD4 and CD8 suppressor T cells; Lymphocytes were delineated by forward/side scatter gating. CD3 was used to identify T cells among the previously selected viable lymphocytes. CD4 T cells and CD8 T cells were identified as uniquely expressing CD4 or CD8 antigens. Tregs were defined as CD4 T cells co-expressing CD25 and FOXP
- FIG. 4C FoxP3 mean fluorescent intensity in CD4 + FOXP3 + CD25 hlgh cell population was comparable among the three groups.
- FIG. 4D CD25 mean fluorescent intensity in the CD4 + FOXP3 + CD25 hlgh cell population was reduced in Alzheimer dementia stage.
- FIG. 4E and FIG. 4F The percentages of CD8 + CD25 hlgh and CD8 + FOXP + suppressor T cells (% of total CD8) were increased in MCI patients, compared to HCs. CD8 + CD25 + T cell population differs significantly between MCI and Alzheimer. P- values are *p ⁇ 0.05 and **p ⁇ 0.01. [0068] Figs. 5A and 5B show the Treg suppressive function on Tresp proliferation.
- FIG. 5A No correlation between age and suppressive activity of Tregs on Tresp proliferation (1:1 ratio) was noted.
- FIG. 5B Suppression (%) of Tregs on Tresp proliferation in MCI us. HC in 1:1 and 2:1 Tresps: Tregs ratios were comparable. Suppression of Tregs on Tresps proliferation in Alzheimer disease in both 1 : 1 and 2: 1 Tresps:Tregs ratios were reduced compared to both MCI and HC.
- P- values are *p ⁇ 0.05, **p ⁇ 0.01, and ***p ⁇ 0.001.
- FIG. 6A-6C show the Treg immunophenotype and suppressive function following ex vivo expansion.
- CD25 (FIG. 6B) and FoxP3 (FIG. 6C) MFIs in CD4 + CD25 high Treg population were elevated following ex vivo expansion. P-values are *p ⁇ 0.05, **p ⁇ 0.01, and ***p ⁇ 0.001.
- FIG. 7A-7E show the suppression of Tregs on iPSC-derived pro-inflammatory macrophages.
- Baseline and ex vivo expanded CD4 + CD25 hlgh Tregs were added to iPSC- derived pro-inflammatory macrophages (Ml) and relative change (% of Ml-only) of pro- inflammatory cytokines were measured.
- FIG. 7A and FIG. 7B At baseline, a trend toward decreased macrophage IL6 transcript expression was noted following co-culture with baseline HC Tregs, however this trend was not observed from IL-6 protein levels. Expanded Tregs of HC, MCI and Alzheimer’s suppressed Ml - IL6 transcript and protein expressions.
- FIG. 7D show the co-culture of baseline Tregs with Ml did not suppress TNFa transcript and protein levels. Reduction of Ml -TNFa transcript and protein expressions were noted following co-culture with expanded Tregs in all three groups.
- FIG. 7E Baseline Tregs did not attenuate ILIB transcript level. Expanded Tregs of Alzheimer disease, MCI and HC displayed an enhanced capacity to suppress Ml-derived ILIB transcript. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001 vs. their corresponding baseline Tregs- Ml co-cultures; ## p ⁇ 0.01, ### p ⁇ 0.001 vs. Ml -alone cultures.
- FIG. 8A and FIG. 8B show the transcript expression profile of immunoregulatory genes in Tregs.
- the transcript levels of IL-13, CD25 and proximity or contact mediated immunoregulatory markers (PD1, GZMB and CD73) were up regulated following ex vivo expansion.
- P-values are *p ⁇ 0.05, **p ⁇ 0.01, and ***p ⁇ 0.001 vs. their corresponding baseline Tregs.
- FIGs. 9A-9H show protein expression of immunoregulatory genes in Tregs.
- protein expression of immunoregulatory genes CD73, PD1, IL-13 and GZMB
- CD73, PD1, IL-13 and GZMB immunoregulatory genes
- FIG. 9A CD73 +
- FIG. 9B PDl +
- FIG. 9C IL13 + Tregs (% of total CD4 + CD25 hlgh Tregs) were noted following ex vivo expansion in both Alzheimer patients and HCs.
- FIG. 10A and FIG. 10B show the suppressive mechanism of ex vivo expanded Tregs.
- Figs. 12A and 12B show the granularity and size, respectively, of expanded and freshly isolated Tregs. These data indicate that expanded Tregs are larger than freshly isolated ones and that expanded Tregs have more granularity than freshly isolated ones.
- Fig. 14 Process flow diagram for the process of Treg isolation and expansion for patients with Amyotrophic Lateral Sclerosis (ALS).
- ALS Amyotrophic Lateral Sclerosis
- Fig. 15 Assembling GE Healthcare-Biosafe CS-490.1 kit (PeriCell).
- Fig. 16 GE Healthcare Biosafe CS-900.2 kit.
- Fig. 17 CliniMACS Tubing Set LS (162-01).
- Fig. 18 CliniMACS® Plus Instrument.
- Fig. 19 Expansion curves for 12 populations of Tregs.
- Fig. 20 shows Viability of Cryopreserved Treg Products.
- Graph shows results for three validation runs.
- Fig. 21 shows Purity of Final Treg Products from FDA Validation Runs. Graph shows results for three validation runs.
- Fig. 22 shows Potency of Cryopreserved Treg Products. Graph shows results for three validation runs. Missing bars indiciate no data could be collected.
- Fig. 23 shows Viability of Final Treg Products from Phase 2a Clinical Trial. Groups of bars show, starting from the, results for Subject Nos. 701-115, 701-114, 701-103, 702-206, 702-205, 702-204, 702-203, 702-202 and 702-201.
- Fig. 24 shows Purity of Final Treg Products from Phase 2a Clinical Trial. Groups of bars show, starting from the, results for Subject Nos. 701-115, 701-114, 701-103, 702-206, 702-205, 702-204, 702-203, 702-202 and 702-201.
- Fig. 25 shows the potency of Final Treg Products from Phase 2a Clinical Trial.
- Graph shows, starting from the, results for Subject Nos. 701-115, 701-114, 701-103, 702- 206, 702-205, 702-204, 702-203, 702-202 and 702-201.
- Fig. 26 shows lack of suppression of proinflammatory macrophages freshly isolated Tregs in vitro. Conditions listed are depicted in the graph from left to right for the Strong Ml (left group of bars) and Weak Ml (right group of bars) experiments.
- Fig. 27 shows suppression of proinflammatory macrophages by expanded/cryopreserved Tregs in vitro. Numbers show percentage decrease compared to No Treg control; * indicates a p-value of 0.05 or less, ** indicates a p value of 0.01 or less, *** indicates a p-value of 0.001 or less.
- Fig. 28 shows a heat map of the results of a proteomics analysis of ALS baseline Tregs, a cryopreserved therapeutic population of Tregs from ALS patients following thawing, and of the Tregs following expansion but prior to cryopreservation.
- Fig. 29 shows a schematic representation of the methods employed in Example 10.
- Fig. 30 shows Nucleofection of GFP-mRNA in T-cells.
- Figs. 31A-31C show Bioinformatics analysis and in-vitro validation of stable mRNA transcripts in T-cells.
- Figs. 32A and 32B show increased cell number and telomere length with hTERT expression in non-transduced and CAR-transduced Tcells. Bars in Fig. 32A and Dots in Fig. 32B show results for Day 0, Day 1 and Day 2 from left to right within each group of three. [0096] Figs. 33A and 33B show nucleofection of GFP and X-mRNA in T regulatory cells (Tregs).
- Fig. 34 shows a summary of the mRNA therapy for improved adoptive T-cell transfer described in Example 10.
- Fig. 35 shows a flow chart of an exemplary process of producing a therapeutic population of Tregs in a bioreactor.
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, and wherein the expansion step begins within about 30-90 minutes of completion of the enrichment step.
- Tregs regulatory T cells
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, wherein the expansion step begins within about 30-90 minutes of completion of the enrichment step, and wherein the cryopreservation step is initiated after about 15-25 days of expansion.
- Tregs regulatory T cells
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, and wherein the expansion step (i) begins within about 30-90 minutes of completion of the enrichment step, (ii) comprises culturing the Tregs in a culture medium that comprises beads coated with anti-CD3 antibodies and anti-CD28 antibodies, and (iii) comprises the addition of an expansion agent to the
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, and wherein the expansion step (i) begins within about 30-90 minutes of completion of the enrichment step, (ii) comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies to the cell culture medium within 24h of initiating the culturing and (iii) comprises the addition of an expansion agent to
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, and wherein the expansion step (i) begins within about 30-90 minutes of completion of the enrichment step, (ii) comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies to the cell culture medium within about 24h of initiating the culturing and (iii) comprises the addition of an expansion agent
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, and wherein the expansion step (i) begins within about 30-90 minutes of completion of the enrichment step, (ii) comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies to the cell culture medium within about 24h of initiating the culturing and (iii) comprises the addition of an expansion agent
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, and wherein the expansion step (i) begins within about 30-90 minutes of completion of the enrichment step, (ii) comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies to the cell culture medium within about 24h of initiating the culturing and (iii) comprises the addition of IL-2
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, and wherein the expansion step (i) begins within about 30-90 minutes of completion of the enrichment step, (ii) comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies to the cell culture medium within about 24h of initiating the culturing, (iii) comprises the addition of an expansion agent
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of (a) enriching Tregs from a leukapheresis sample suspected of containing Tregs, to produce a baseline Treg cell population; (b) expanding the baseline Treg cell population to produce an expanded Treg cell population, wherein the baseline Treg cell population is not cryopreserved prior to the initiation of step (b); and (c) cryopreserving the expanded Treg cell population to produce a cryopreserved therapeutic population of Tregs, wherein the enriching step begins within about 30 to 90 minutes of obtaining the leukapheresis sample, wherein the expansion step (i) begins within about 30-90 minutes of completion of the enrichment step, (ii) comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies to the cell culture medium within about 24h of initiating the culturing, (iii) comprises the addition of an expansion agent to
- FIG. 1 A detailed overview of an exemplary manufacturing process is depicted in FIG.
- Tregs When isolating Tregs for clinical therapies, a CD8/CD19 depletion step followed by a CD25 enrichment step is generally employed and subsequent ex vivo expansion in the presence of IL-2, which supports the survival and proliferation of Tregs, and rapamycin, which stabilizes the Treg population.
- This isolation and expansion strategy reduces the amount of pro-inflammatory populations (i. e., cytotoxic CDS+ T cells and T effector cells), and increases the purity of the final Treg product.
- the robust manufacturing of Tregs is limited by the low number of circulating Tregs that can be isolated through leukapheresis and ex vivo separation. Hence, extensive ex vivo expansion is essential in order to obtain sufficient numbers of Tregs to treat patients for a longer period of time.
- the expansion of functionally compromised Tregs for example, autologous ALS-derived Tregs, is even more challenging.
- Amyotrophic lateral sclerosis also known as Lou Gehrig’s disease
- Lou Gehrig is a rapidly progressive and fatal neurodegenerative disease characterized by the relentless degeneration of upper and lower motor neurons.
- Increasing evidence shows that dysregulation of the immune system can hasten ALS disease progression.
- Tregs are reduced in patients with ALS and more marked reduction is associated with more rapid disease progression.
- Tregs are a subpopulation of T-lymphocytes consisting of CD4 D25 1 ' 1 hFOXP3 + cells that suppress neuroinflammatory responses. This work is the first to identify Treg compositions as a therapy for patients with neurological disorders such as ALS.
- the Treg manufacturing processes described herein overcome challenges of robust Treg expansion and cryopreservation while maintaining phenotypic characteristics and functionality.
- Current Treg manufacturing protocols are complicated and labor-intensive, which drive up manufacturing costs and are not sustainable as a therapy for large numbers of patients, for example patients with neurodegenerative diseases such as ALS or Alzheimer’s disease.
- the methods described herein address manufacturing challenges at a lower cost through optimization of the manufacturing process, e.g., cGMP manufacturing processes.
- the methods described herein produce an enhanced Treg product with superior suppressive functions that are maintained even when the Tregs are cryopreserved and thawed without further expansion, wherein the Tregs could be more efficacious when infused back into the patients.
- the improved Treg manufacturing processes described herein are critical to the advancement of developing a therapy that drastically slows disease progression in ALS because it provides a platform for current and future clinical studies in ALS, and for therapeutic ALS regimens, allows for the effective cryopreservation of ALS-derived autologous Tregs for extended treatment times with successive doses, generates functionally superior Treg products, and is potentially an “off-the-shelf immune- privileged Treg therapy that can be used for treatment of diseases, for example, neurodegenerative diseases including ALS and Alzheimer’s disease, autoimmune diseases including Type 1 diabetes and rheumatoid arthritis, and graft versus host disease (GVHD) including GVHD following bone marrow transplantation.
- diseases for example, neurodegenerative diseases including ALS and Alzheimer’s disease, autoimmune diseases including Type 1 diabetes and rheumatoid arthritis, and graft versus host disease (GVHD) including GVHD following bone marrow transplantation.
- diseases for example, neurodegenerative diseases including ALS and Alzheimer’s
- Tregs Regulatory T cells
- ALS amyotrophic lateral sclerosis
- IL-2 interleukin-2
- rapamycin rapamycin
- expanded functional Tregs represent a potential treatment for ALS that could slow the rate of progression.
- the particular susceptibility of the Treg population to the ongoing disease process and the autologous nature of the proposed treatment pose significant challenges to the development of a Treg therapy that could combat ALS in potentially thousands of patients.
- a sensible Treg manufacturing process for ALS patients requires an expansion phase that yields sufficient numbers of highly suppressive Tregs to avoid exposing the patients to frequent leukapheresis procedures for Treg isolation. Frequent infusions of optimized Treg doses would be required to continually suppress the progressive neuroinflammatory environment that ensues as ALS progresses. As it would be impractical to expand Tregs from each ALS patient prior to each infusion, the development of a cryopreservation process is crucial in order to limit the per-patient costs of cell manufacturing as well as the man power that would be needed to develop Treg therapies for potentially thousands of patients with ALS.
- Treg manufacturing processes described herein have been optimized to produce and cryopreserve large numbers of highly suppressive Tregs for their application in treatment regimens, for example, for their application in methods of treating disorders such as, e.g., neurodegenerative disorders such as ALS and Alzheimer’s disease, autoimmune disorders such as Type 1 diabetes and rheumatoid arthritis, and graft versus host disease (GVHD) such as following a bone marrow transplantation, as well as for their application in future clinical trials for ALS patients and potentially other neurodegenerative diseases such as Alzheimer’s disease.
- An optimized Treg therapy minimizes the number of expansion phases and infusions, which makes the therapy more cost effective and sustainable.
- a method of producing a cryopreserved therapeutic population of regulatory T cells comprising the steps of:
- Treg-enriched cell population (baseline Treg cell population);
- step (b) expanding the Treg-enriched cell population to produce an expanded Treg cell population, wherein the Treg-enriched cell population is not cryopreserved prior to the initiation of step (b);
- step (a) comprises enriching for CD25 + cells.
- step (a) comprises depleting CD8 + /CD19 + cells.
- step (a) comprises depleting CD8+/CD19+ cells then enriching for CD25+ cells.
- step (b) is carried out within 30 minutes after step (a).
- step (b) comprises culturing the Tregs in a culture medium that comprises at least one expansion agent.
- the expansion agent is a CD3 -activating agent.
- the CD3 -activating agent is an anti-CD3 antibody.
- the CD3 -activating agent is first added to the culture medium within about 24 hours of the initiation of the culturing.
- step (b) further comprises culturing in a culture medium that comprises a CD3 -activating agent.
- the CD3 -activating agent is an anti-CD3 antibody.
- the CD3 -activating agent is first added to the culture medium within about 24 hours of the initiation of the culturing.
- step (b) comprises culturing the Tregs in a culture medium that comprises beads present in a beadxell ratio of 4: 1, wherein the beads are coated with anti-CD3 antibodies and anti-CD28 antibodies.
- the method of embodiment 6 wherein the expansion agent is IL-2.
- step (b) further comprises replenishing the culture medium with IL-2 about every 2-3 days after IL-2 was first added to the culture medium.
- step (b) further comprises culturing in a culture medium that comprises a CD3 -activating agent.
- the CD3 -activating agent is an anti-CD3 antibody.
- step (b) further comprises culturing in a culture medium that comprises a CD28-activating agent.
- step (b) further comprises culturing in a culture medium that comprises a CD3 -activating agent.
- step (b) comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies to the culture medium, in a beadxell ratio of 4: 1, within about 24 hours of the initiation of culturing; adding the IL-2 to the culture medium within about 6 days of the initiation of culturing; and replenishing the culture medium with IL-2 every 2-3 days after the IL-2 is first added to the culture medium.
- step (b) further comprises adding beads coated with anti-CD3 antibodies and anti-CD28 antibodies, in a beadxell ratio of 1:1, wherein the adding occurs about 14 days after beads coated with anti-CD3 antibodies and anti-CD28 antibodies were first added to the culture medium.
- step (b) comprises culturing in a culture medium that comprises a mammalian target of rapamycin (mTor) inhibitor.
- mTor mammalian target of rapamycin
- the method of embodiment 36 or embodiment 37 further comprises replenishing the culture medium with rapamycin every 2-3 days after the rapamycin is first added to the culture medium.
- the cryopreserving step (c) is carried out at least 6 days following an addition of IL-2 to the culture medium in step (b).
- cryopreserving step (c) is carried out about 12-25 days after the initiation of the culturing step (b).
- step(c) comprises cryopreserving the Tregs in a cryoprotectant comprising DMSO.
- the cryoprotectant comprises 10% DMSO.
- cryopreservation step (c) comprises changing the temperature of the population of Tregs in the following increments: l°C/min to 4°C, 25°C/min to -40°C, 10°C/min to -12°C, l°C/min to -40°C, and 10°C/min to -80°C-90°C.
- the method of any one of embodiments 1-43 wherein the cryopreserved therapeutic population of Tregs is stored in liquid nitrogen vapor phase.
- the cryopreserved therapeutic population of Tregs is frozen at a Treg density of at least 50 million cells. mL.
- any one of embodiments 1-45 wherein the cryopreserved therapeutic population of Tregs is frozen at a concentration of lxlO 6 cells per kg of body weight of an intended recipient of the Tregs per mL.
- cryopreserved therapeutic population of Tregs is frozen in a total volume of 1-1.5 mL.
- the method further comprises thawing the population of Tregs after cryopreservation for about 1 week, 1 month, about 3 months, about 6 months, about 9 months, about 12 months, 18 months or about 24 months.
- the method of embodiment 50 wherein the cryopreserved therapeutic population of Tregs is thawed in 50mL of a solution comprising 0.9% sodium chloride and about 5% human serum albumin.
- the method of embodiment 50 wherein the therapeutic population of Tregs was cryopreserved in a single cryovial containing lxlO 6 Tregs per kg of body weight of an intended recipient of the Tregs per mL and is thawed in 50 mL of a solution containing 0.9% sodium chloride and about 5% human serum albumin.
- the method of embodiment 52 wherein the therapeutic population of Tregs in the single cryovial is contained in a volume of 1-1.5 mL.
- the method of any one of embodiments 1-53 wherein the method further comprises obtaining the cell sample from a donor by leukapheresis.
- the method of embodiment 57, wherein the human donor is diagnosed with or is suspected of having a disorder associated with Treg deficiency.
- the neurodegenerative disorder is Amyotrophic Lateral Sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease, Huntington’s disease or frontotemporal dementia.
- ALS Amyotrophic Lateral Sclerosis
- the cell sample is from a donor diagnosed with or suspected of having an autoimmune disease.
- the autoimmune disease is systemic sclerosis (scleroderma), polymyosititis, ulcerative colitis, inflammatory bowel disease, Crohn’s disease, celiac disease, multiple sclerosis (MS), rheumatoid arthritis (RA), Type I diabetes, psoriasis, dermatomyostitis, systemic lupus erythematosus, cutaneous lupus, myasthenia gravis, autoimmune nephropathy, autoimmune hemolytic anemia, autoimmune cytopenia autoimmune hepatitis, autoimmune uveititis, alopecia, thyroiditis or pemhigus.
- systemic sclerosis systemic sclerosis
- polymyosititis ulcerative colitis
- inflammatory bowel disease Crohn’s disease
- MS multiple sclerosis
- RA rheumatoid arthritis
- Type I diabetes psoriasis, dermatomyostitis, systemic l
- the method of embodiment 57 wherein the human donor is diagnosed with or suspected of having heart failure or ischemic cardiomyopathy.
- the method of embodiment 66, wherein the genetic engineering comprises the introduction of a transgene into the Tregs.
- the method of embodiment 66, wherein the genetic engineering comprises the introduction of an mRNA into the Tregs.
- the method of embodiment 66, wherein the genetic engineering comprises the gene editing through CRISPR-Cas9.
- the method of embodiment 66, wherein the genetic engineering comprises the reduction of gene expression through siRNA or antisense oligonucleotides.
- step (b) is automated.
- step (b) takes place in a bioreactor.
- step (b) takes place in a G- REX culture system.
- the method is performed in a closed system.
- the method of embodiment 1, wherein the Treg-enriched cell population comprises dysfunctional Tregs.
- cryopreserved therapeutic population of Tregs comprises an increased proportion of O ⁇ 4 + O ⁇ 25 w ⁇ 1 Tregs relative to the proportion of O ⁇ 4 + O ⁇ 25 M ⁇ 1 Tregs in the Treg- enriched cell population, as determined by flow cytometry.
- cryopreserved therapeutic population of Tregs comprises an increased proportion of CD4 + CD25 high CD127 low Tregs relative to the proportion of CD4+CD25 high CD127 low Tregs in the Treg- enriched cell population, as determined by flow cytometry.
- cryopreserved therapeutic population of Tregs comprises CD25 + Tregs wherein the expression of CD25 in the Tregs is increased relative to the expression of CD25 in the Tregs in the Treg- enriched cell population, as determined by flow cytometry.
- cryopreserved population of Tregs comprises CD127 + Tregs wherein the expression of CD127 in the Tregs is not increased greater than 3-fold relative to the expression of CD127 in the Tregs in the Treg-enriched cell population, as determined by flow cytometry.
- any one of embodiments 1-81 wherein the granularity of the Tregs in the cryopreserved therapeutic population of Tregs is increased relative to the granularity of the Tregs in the Treg- enriched cell population, as determined by flow cytometry.
- the method of embodiment 82 wherein the granularity of the Tregs increased by at least about 1.5-fold, at least about 2-fold, or at least about 2.5-fold.
- the size of the Tregs increased by at least about 1.2-fold, at least about 1.5-fold, or at least about 2-fold.
- the Tregs in the cryopreserved therapeutic population of Tregs comprise CTLA4-expressing Tregs wherein the expression of CTLA4 is increased in the Tregs relative to expression of CTLA4 in the Tregs in the Treg-enriched cell population.
- the cryopreserved therapeutic population of Tregs comprises CTLA4+ Tregs wherein the proportion of CTLA4+ Tregs is increased relative to the proportion of CTLA4+ Tregs in the Tregs in the Treg-enriched cell population.
- cryopreserved therapeutic population of Tregs comprises at least 20%, at least 30%, at least 40%, or at least 50% CTLA4+ Tregs, as determined by flow cytometry.
- cryopreserved therapeutic population of Tregs comprises FoxP3+ Tregs wherein the proportion of FoxP3+ Tregs is increased relative to the proportion of FoxP3+ Tregs in the Tregs in the Treg- enriched cell population.
- cryopreserved therapeutic population of Tregs comprises at least 40%, at least 50%, at least 60%, at least 70%, or at least 80% or at least 90% FoxP3+ Tregs, as determined by flow cytometry.
- the Tregs in the cryopreserved therapeutic population of Tregs comprise FoxP3 -expressing Tregs wherein the expression of FoxP3 is increased in the Tregs relative to expression of FoxP3 in the Tregs in the Treg-enriched cell population.
- cryopreserved therapeutic population of Tregs comprises at least 70%, at least 80%, or at least 90% CD4 + CD25 high CD127 low Tregs in the Treg-enriched cell population, as determined by flow cytometry.
- cryopreserved therapeutic population of Tregs comprises at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% CD4 + CD25 + cells, as determined by flow cytometry.
- the cryopreserved population of Tregs comprise fewer than 20% CD8+ cells, determined by flow cytometry.
- the viability of the cryopreserved therapeutic population of Tregs is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the viability of the expanded Treg cell population prior to the expanded Treg cell population being cryopreserved in step (c).
- any one of embodiments 1-96 wherein the suppressive function of the cryopreserved population of Tregs is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%, as determined by flow cytometry performed following thawing of the therapeutic population.
- the method of any one of embodiments 1-97 wherein the cryopreserved population of Tregs exhibits a suppressive function that is greater than the suppressive function of the enriched Treg cell population, as measured prior to step (b)), as measured by a Treg suppression assay.
- the suppressive function of the cryopreserved therapeutic population of Tregs is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the suppressive function of the expanded Treg cell population prior to the Tregs being cryopreserved in step (c), as determined by flow cytometry. .
- cryopreserved therapeutic population of Tregs following thawing, exhibits an ability to suppress inflammatory cells, as measured by IL-6 production by the inflammatory , wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells.
- a composition comprising a cryopreserved therapeutic population of Tregs produced by the method of any one of embodiments 1-100.
- a cryopreserved composition comprising a therapeutic population of Tregs, wherein upon thawing the therapeutic population of Tregs exhibits greater 50% suppressive function in the absence of need for additional expansion. .
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing the therapeutic population of Tregs exhibits greater than 50% suppressive function in the absence of need for additional expansion.
- cryopreserved composition of any one of embodiments 102-104 wherein the suppressive function of the therapeutic population of Tregs following thawing of the cryopreserved composition is at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the suppressive function of the Tregs before undergoing cryopreservation, as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation. .
- composition of any one of embodiments 102-105 wherein upon thawing, the therapeutic population of Tregs exhibits at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% viability, as determined by trypan blue staining.
- composition of any one of embodiments 102-106 wherein upon thawing, the therapeutic population of Tregs exhibits a viability that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the viability of the Tregs before undergoing cryopreservation, as determined by trypan blue staining..
- cryopreserved composition of any one of embodiments 103-114 wherein upon thawing, the expression of one or more gene products listed in Table 14 is increased relative to the expression of the one or more gene products in baseline Tregs prior to expansion.
- cryopreserved composition of embodiment 118 wherein the expression of one or more of the following gene products is increased in the therapeutic population of Tregs relative to the expression of the one or more gene products in baseline Tregs prior to expansion: ADAM 10, AIMP1, AIMP2, ARG2, BCL2L1, BSG, CD2, CD28, CD38, CD74, CD84, CTLA4, FAS, FOXP3, GCLC, HAT1, HLA-DQA1, HLA- DQB1, HLA-DRA, HLA-DRB1, HPGD, ICOS, ILIRN, IRF4, KPNA2, LGALS1, LGMN, PCNA, POFUT1, SATB1, SELPLG, STAT1, TFRC, TNFRSF18, .
- cryopreserved composition of any one of embodiments 103-114 wherein upon thawing, the expression of one or more gene products listed in Table 16 is increased relative to the expression of the one or more gene products in baseline Tregs prior to expansion.
- the cryopreserved composition of embodiment 120 wherein the expression of one or more of the following gene products is increased in the therapeutic population of Tregs relative to the expression of the one or more gene products in baseline Tregs prior to expansion: CAA2, ACADM, ACADVL, ACOT7,ACSLl, ACSL4, ACSL5, AGK, AGMAT, AK4, ARG2, ARL2, AUH, BCL2L1, BDH1, BNIP1, CDK1,
- cryopreserved composition of embodiment 122 wherein the expression of one or more of the following gene products is increased in the therapeutic population of Tregs relative to the expression of the one or more gene products in baseline Tregs prior to expansion: ARL2, ARL3, BCCIP, CCDC124, CDK1, CDK2, CDK5, CDK6, CUL4B, DCTN3, FEN1, HELLS, LIG1, MAD2L1, MAEA, MCM2, MCM2, MCM3, MCM4, MCM5, MCM6, MCM7, MCMBP, NUDC, PCNA, POLD1, POLD2, RALB, RBM38, RFC2, RFC3, RFC4, RFC5, RNASEH2A, RNASEH2B, and SMC2. .
- cryopreserved composition of any one of embodiments 116-125 wherein the expression is changed at least about 4-fold, at least about 5-fold, at least about 6- fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, or at least about 10-fold.
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing, the expression of one or more of the geneproducts listed in Tables 6-11 is substantially the same in the therapeutic population of Tregs as the expression of the one or more gene products in the Tregs prior to cryopreservation.
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing, the expression of one or more gene products listed in Table 12 and/or Table 13 is decreased relative to the expression of the one or more gene products in baseline Tregs prior to expansion. .
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing, the expression of one or more gene products listed in Table 14 is increased relative to the expression of the one or more gene products in baseline Tregs prior to expansion.
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing, the expression of one or more gene products listed in Table 15 is increased relative to the expression of the one or more gene products in baseline Tregs prior to expansion.
- cryopreserved composition of embodiment 131 wherein the expression of one or more of the following gene products is increased in the therapeutic population of ex v/vo-expanded Tregs relative to the expression of the one or more gene products in baseline Tregs prior to expansion: ADAMIO, AIMP1, AIMP2, ARG2, BCL2L1, BSG, CD2, CD28, CD38, CD74, CD84, CTLA4, FAS, FOXP3, GCLC, HAT1, HLA- DQA1, HLA-DQB1, HLA-DRA, HLA-DRB1, HPGD, ICOS, IL1RN, IRF4, KPNA2, LGALSl, LGMN, PCNA, POFUT1, SATB1, SELPLG, STAT1, TFRC, TNFRSF18,.
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing, the expression of one or more gene products listed in Table 16 is increased relative to the expression of the one or more gene products in baseline Tregs prior to expansion.
- the cryopreserved composition of embodiment 133 wherein the expression of one or more of the following gene products is increased in the therapeutic population of Tregs relative to the expression of the one or more gene products in baseline Tregs prior to expansion: CAA2, ACADM, ACADVL, ACOT7,ACSLl, ACSL4, ACSL5, AGK, AGMAT, AK4, ARG2, ARL2, AUH, BCL2L1, BDH1, BNIP1, CDK1,
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing, the expression of one or more gene products listed in Table 17 is increased relative to the expression of the one or more gene products in baseline Tregs prior to expansion.
- a cryopreserved composition comprising a therapeutic population of ex vivo- expanded Tregs, wherein upon thawing, the expression of one or more gene products listed in Table 18 is increased relative to the expression of the one or more gene products in baseline Tregs prior to expansion.
- cryopreserved composition of embodiment 137 wherein the expression of one or more of the following gene products is increased in the therapeutic population of Tregs relative to the expression of the one or more gene products in baseline Tregs prior to expansion: ACAA2, ACADM, ACADVL, ACOT7, BSG, CACYBP, CD74, CDK1, CPOX, DUT, ECU, EN03, FEN1, FKBP3, HIST1H2BJ, HLA-DQA1, HLA- DRA, HLA-DRBl, LGALS1, LGALS3, MCM5, MCM6, MCM7, MTHFD1, NAMPT, NME1, NQOl, PCNA, RABIA, RALB, SLC25A4, STAT1, STMN1, STMN2, TUBA1B, TUBB4A, TUBB8, TXN, TXNRD1, and WARS.
- cryopreserved composition of any one of embodiments 129-137 wherein the expression is changed at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, or at least about 10- fold.
- cryopreserved composition of any one of embodiments 102-140 wherein the cryopreserved therapeutic population of Tregs, following thawing, exhibits an ability to suppress inflammatory cells, as measured by IL-6 production by the inflammatory cells, wherein the inflammatory cells are macrophages or monocytes from human donors or generated from induced pluripotent stem cells.
- the cryopreserved composition of any one of embodiments 102-140 wherein the cryopreserved composition comprises DMSO.
- the cryopreserved composition of embodiment 141, wherein the cryopreserved composition comprises 10% DMSO. .
- the cryopreserved composition any one of embodiments 102-142, wherein the therapeutic population of Tregs is present at a concentration of lxlO 6 cells per kg of body weight of an intended recipient of the Treg population per ml. .
- the cryopreserved composition any one of embodiments 102-144, wherein the cryopreserved composition is contained a single cryovial. .
- the cryopreserved composition any one of embodiments 102-145, wherein the total volume of the cryopreserved composition is 1-1.5 mL. .
- a method of treating a disorder associated with Treg dysfunction comprising: administering to a subject in need of said treatment a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering. .
- a method of treating a disorder associated with Treg deficiency comprising: administering to a subject in need of said treatment a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating a disorder associated with overstimulation of the immune system comprising: administering to a subject in need of said treatment a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering. .
- a method of treating an inflammatory condition driven by a T cell response comprising: administering to a subject in need of said treatment a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating a neurodegenerative disorder in a subject in need thereof comprising: administering to the subject a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the neurodegenerative disease is Amyotrophic Lateral Sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease, frontotemporal dementia or Huntington’s disease.
- ALS Amyotrophic Lateral Sclerosis
- the autoimmune disorder is polymyosititis, ulcerative colitis, inflammatory bowel disease, Crohn’s disease, celiac disease, multiple sclerosis (MS), rheumatoid arthritis (RA), Type I diabetes, psoriasis, dermatomy ostitis, systemic lupus erythematosus, cutaneous lupus, myasthenia gravis, autoimmune nephropathy, autoimmune hemolytic anemia, autoimmune cytopenia autoimmune hepatitis, autoimmune uveititis, alopecia, thyroiditis or pemhigus. .
- a method of treating graft-versus-host disease in a subject in need thereof comprising: administering to the subject a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the method of embodiment 157 wherein the subject has received a bone marrow transplant, kidney transplant or liver transplant.
- a method of improving islet graft survival in a subject in need thereof comprising: combining islet transplantation with administering to the subject a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering. .
- a method of treating cardio-inflammation in a subject in need thereof comprising: administering to the subject a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- the method of embodiment 160 wherein the cardio-inflammation is associated with a atherosclerosis, myocardial infarction, ischemic cardiomyopathy or heart failure.
- a method of treating neuroinflammation in a subject in need thereof comprising: administering to the subject a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering.
- a method of treating a Tregopathy in a subject in need thereof comprising: administering to the subject a therapeutic population of Tregs, wherein the Tregs had been ex vivo expanded and cryopreserved, and wherein the Tregs are not further expanded prior to the administering. .
- CTLA4 cytotoxic T lymphocyte-associated antigen 4
- LRBA LPS- responsive and beige-like anchor protein
- BACH2 BTB domain and CNC homolog 2
- STAT3 signal transducer and activator of transcription 3
- any one of embodiments 149-167 wherein, the therapeutic population of Tregs is administered to the subject in an aqueous solution comprising 0.9% sodium chloride and 5% human serum albumin.
- a method of treating a neurodegenerative disorder in a subject in need thereof comprising: administering to the subject a pharmaceutical composition comprising a therapeutic population of Tregs, wherein the pharmaceutical composition comprises the cryopreserved composition of any one of embodiments 102-148, thawed and suspended in an aqueous solution suitable for administration to the subject.
- the method of embodiment 168, wherein the neurodegenerative disease is Amyotrophic Lateral Sclerosis (ALS), Alzheimer’s disease or Parkinson’s disease..
- ALS Amyotrophic Lateral Sclerosis
- aqueous solution comprises 0.9% sodium chloride and 5% human serum albumin.
- the therapeutic population of Tregs are administered within 6 hours of thawing.
- any one of embodiments 138-171 wherein about lxlO 6 Tregs per kg of body weight of the subject are administered per month for the first and second month, and about 2xl0 6 Tregs per kg of body weight of the subject are administered per month for at least a third and fourth month.
- polynucleotides, nucleic acid segments, nucleic acid sequences, and the like include, but are not limited to, DNAs (including and not limited to genomic or extragenomic DNAs), genes, peptide nucleic acids (PNAs) RNAs (including, but not limited to, rRNAs, mRNAs and tRNAs), nucleosides, and suitable nucleic acid segments either obtained from natural sources, chemically synthesized, modified, or otherwise prepared or synthesized in whole or in part by the hand of man.
- DNAs including and not limited to genomic or extragenomic DNAs
- genes include peptide nucleic acids (PNAs) RNAs (including, but not limited to, rRNAs, mRNAs and tRNAs), nucleosides, and suitable nucleic acid segments either obtained from natural sources, chemically synthesized, modified, or otherwise prepared or synthesized in whole or in part by the hand of man.
- PNAs peptide nucleic acids
- Biocompatible refers to a material that, when exposed to living cells, will support an appropriate cellular activity of the cells without causing an undesirable effect in the cells, such as a change in a living cycle of the cells, a change in a proliferation rate of the cells, or a cytotoxic effect.
- biologically-functional equivalent is well understood in the art, and is further defined in detail herein. Accordingly, sequences that have about 85% to about 90%; or more preferably, about 91% to about 95%; or even more preferably, about 96% to about 99%; of nucleotides that are identical or functionally-equivalent to one or more of the nucleotide sequences provided herein are particularly contemplated to be useful in the practice of the methods and compositions set forth in the instant application.
- biomimetic shall mean a resemblance of a synthesized material to a substance that occurs naturally in a human body and which is not rejected by (e.g, does not cause an adverse reaction in) the human body.
- buffer includes one or more compositions, or aqueous solutions thereof, that resist fluctuation in the pH when an acid or an alkali is added to the solution or composition that includes the buffer. This resistance to pH change is due to the buffering properties of such solutions, and may be a function of one or more specific compounds included in the composition. Thus, solutions or other compositions exhibiting buffering activity are referred to as buffers or buffer solutions. Buffers generally do not have an unlimited ability to maintain the pH of a solution or composition; rather, they are typically able to maintain the pH within certain ranges, for example from a pH of about 5 to 7.
- carrier is intended to include any solvent(s), dispersion medium, coating(s), diluent(s), buffer(s), isotonic agent(s), solution(s), suspension(s), colloid(s), inert(s) or such like, or a combination thereof, that is pharmaceutically acceptable for administration to the relevant animal.
- delivery vehicles for chemical compounds in general, and chemotherapeutics in particular is well known to those of ordinary skill in the pharmaceutical arts. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the diagnostic, prophylactic, and therapeutic compositions is contemplated.
- One or more supplementary active ingredient(s) may also be incorporated into, or administered in association with, one or more of the disclosed chemotherapeutic compositions.
- DNA segment refers to a DNA molecule that has been isolated free of total genomic DNA of a particular species. Therefore, a DNA segment obtained from a biological sample using one of the compositions disclosed herein refers to one or more DNA segments that have been isolated away from, or purified free from, total genomic DNA of the particular species from which they are obtained. Included within the term “DNA segment,” are DNA segments and smaller fragments of such segments, as well as recombinant vectors, including, for example, plasmids, cosmids, phage, viruses, and the like.
- the term “effective amount,” as used herein, refers to an amount that is capable of treating or ameliorating a disease or condition or otherwise capable of producing an intended therapeutic effect.
- a heterologous sequence is defined in relation to a predetermined, reference sequence, such as, a polynucleotide or a polypeptide sequence.
- a heterologous promoter is defined as a promoter which does not naturally occur adjacent to the referenced structural gene, but which is positioned by laboratory manipulation.
- a heterologous gene or nucleic acid segment is defined as a gene or segment that does not naturally occur adjacent to the referenced promoter and/or enhancer elements.
- homologous means, when referring to polynucleotides, sequences that have the same essential nucleotide sequence, despite arising from different origins. Typically, homologous nucleic acid sequences are derived from closely related genes or organisms possessing one or more substantially similar genomic sequences. By contrast, an “analogous” polynucleotide is one that shares the same function with a polynucleotide from a different species or organism, but may have a significantly different primary nucleotide sequence that encodes one or more proteins or polypeptides that accomplish similar functions or possess similar biological activity. Analogous polynucleotides may often be derived from two or more organisms that are not closely related (e.g., either genetically or phylogenetically).
- the term “homology” refers to a degree of complementarity between two or more polynucleotide or polypeptide sequences.
- the word “identity” may substitute for the word “homology” when a first nucleic acid or amino acid sequence has the exact same primary sequence as a second nucleic acid or amino acid sequence.
- Sequence homology and sequence identity can be determined by analyzing two or more sequences using algorithms and computer programs known in the art. Such methods may be used to assess whether a given sequence is identical or homologous to another selected sequence.
- nucleic acid or polypeptide sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using one of the sequence comparison algorithms described below (or other algorithms available to persons of ordinary skill) or by visual inspection.
- implantable or “suitable for implantation” means surgically appropriate for insertion into the body of a host, e.g, biocompatible, or having the desired design and physical properties.
- the phrase “in need of treatment” refers to a judgment made by a caregiver such as a physician or veterinarian that a patient requires (or will benefit in one or more ways) from treatment. Such judgment may made based on a variety of factors that are in the realm of a caregiver’s expertise, and may include the knowledge that the patient is ill as the result of a disease state that is treatable by one or more compound or pharmaceutical compositions such as those set forth herein.
- the phrases “isolated” or “biologically pure” refer to material that is substantially, or essentially, free from components that normally accompany the material as it is found in its native state.
- kit may be used to describe variations of the portable, self-contained enclosure that includes at least one set of reagents, components, or pharmaceutically-formulated compositions to conduct one or more of the assay methods of the present invention.
- kit may include one or more sets of instructions for use of the enclosed reagents, such as, for example, in a laboratory or clinical application.
- Link refers to any method known in the art for functionally connecting one or more proteins, peptides, nucleic acids, or polynucleotides, including, without limitation, recombinant fusion, covalent bonding, disulfide bonding, ionic bonding, hydrogen bonding, electrostatic bonding, and the like.
- naturally-occurring refers to the fact that an object can be found in nature.
- a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by the hand of man in a laboratory is naturally-occurring.
- laboratory strains of rodents that may have been selectively bred according to classical genetics are considered naturally- occurring animals.
- nucleic acid includes one or more types of: polydeoxyribonucleotides (containing 2-deoxy-D-ribose), polyribonucleotides (containing D- ribose), and any other type of polynucleotide that is an N-glycoside of a purine or pyrimidine base, or modified purine or pyrimidine bases (including abasic sites).
- nucleic acid also includes polymers of ribonucleosides or deoxyribonucleosides that are covalently bonded, typically by phosphodiester linkages between subunits, but in some cases by phosphorothioates, methylphosphonates, and the like. “Nucleic acids” include single- and double-stranded DNA, as well as single- and double-stranded RNA.
- nucleic acids include, without limitation, gDNA; hnRNA; mRNA; rRNA, tRNA, micro RNA (miRNA), small interfering RNA (siRNA), small nucleolar RNA (snORNA), small nuclear RNA (snRNA), and small temporal RNA (stRNA), and the like, and any combination thereof.
- operably linked and operatively linked refers to that union of the nucleic acid sequences that are linked in such a way, such that the coding regions are contiguous and in correct reading frame. Such sequences are typically contiguous, or substantially contiguous. However, since enhancers generally function when separated from the promoter by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably linked but not contiguous.
- the term “patient” refers to any host that can receive one or more of the pharmaceutical compositions disclosed herein.
- the subject is a vertebrate animal, which is intended to denote any animal species (and preferably, a mammalian species such as a human being).
- a “patient” refers to any animal host including without limitation any mammalian host.
- the term refers to any mammalian host, the latter including but not limited to, human and non-human primates, bovines, canines, caprines, cavines, corvines, epines, equines, felines, hircines, lapines, leporines, lupines, murines, ovines, porcines, ranines, racines, vulpines, and the like, including livestock, zoological specimens, exotics, as well as companion animals, pets, and any animal under the care of a veterinary practitioner.
- a patient can be of any age at which the patient is able to respond to inoculation with the present vaccine by generating an immune response.
- the mammalian patient is preferably human.
- phrases “pharmaceutically-acceptable” refers to molecular entities and compositions that preferably do not produce an allergic or similar untoward reaction when administered to a mammal, and in particular, when administered to a human.
- salts refers to a salt that preferably retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects.
- examples of such salts include, without limitation, acid addition salts formed with inorganic acids (e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like); and salts formed with organic acids including, without limitation, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic (embonic) acid, alginic acid, naphthoic acid, polyglutamic acid, naphthalenesulfonic acids, naphthalenedisulfonic acids, polygalacturonic acid; salts with polyvalent metal cations such as zinc, calcium, bismuth
- plasmid refers to a genetic construct that is composed of genetic material (i.e., nucleic acids).
- a plasmid or a vector contains an origin of replication that is functional in bacterial host cells, e.g., Escherichia coli, and selectable markers for detecting bacterial host cells including the plasmid.
- Plasmids and vectors of the present invention may include one or more genetic elements as described herein arranged such that an inserted coding sequence can be transcribed and translated in a suitable expression cells.
- the plasmid or vector may include one or more nucleic acid segments, genes, promoters, enhancers, activators, multiple cloning regions, or any combination thereof, including segments that are obtained from or derived from one or more natural and/or artificial sources.
- polymer means a chemical compound or mixture of compounds formed by polymerization and including repeating structural units. Polymers may be constructed in multiple forms and compositions or combinations of compositions.
- polypeptide is intended to encompass a singular “polypeptide” as well as plural “polypeptides,” and includes any chain or chains of two or more amino acids.
- terms including, but not limited to “peptide,” “dipeptide,” “tripeptide,” “protein,” “enzyme,” “amino acid chain,” and “contiguous amino acid sequence” are all encompassed within the definition of a “polypeptide,” and the term “polypeptide” can be used instead of, or interchangeably with, any of these terms.
- polypeptides that have undergone one or more post-translational modification(s), including for example, but not limited to, glycosylation, acetylation, phosphorylation, amidation, derivatization, proteolytic cleavage, post-translation processing, or modification by inclusion of one or more non-naturally occurring amino acids.
- post-translational modification(s) including for example, but not limited to, glycosylation, acetylation, phosphorylation, amidation, derivatization, proteolytic cleavage, post-translation processing, or modification by inclusion of one or more non-naturally occurring amino acids.
- Conventional nomenclature exists in the art for polynucleotide and polypeptide structures.
- amino acids Alanine (A; Ala), Arginine (R; Arg), Asparagine (N; Asn), Aspartic Acid (D; Asp), Cysteine (C; Cys), Glutamine (Q; Gin), Glutamic Acid (E; Glu), Glycine (G; Gly), Histidine (H; His), Isoleucine (I; lie), Leucine (L; Leu), Methionine (M; Met), Phenylalanine (F; Phe), Proline (P; Pro), Serine (S; Ser), Threonine (T; Thr), Tryptophan (W; Trp), Tyrosine (Y; Tyr), Valine (V; Vai), and Lysine (K; Lys).
- Amino acid residues described herein are preferred to be in the “1” isomeric form. However, residues in the “d” isomeric form may be substituted for any 1 -amin
- the terms “prevent,” “preventing,” “prevention,” “suppress,” “suppressing,” and “suppression” as used herein refer to administering a compound either alone or as contained in a pharmaceutical composition prior to the onset of clinical symptoms of a disease state so as to prevent any symptom, aspect or characteristic of the disease state. Such preventing and suppressing need not be absolute to be deemed medically useful.
- Protein is used herein interchangeably with “peptide” and “polypeptide,” and includes both peptides and polypeptides produced synthetically, recombinantly, or in vitro and peptides and polypeptides expressed in vivo after nucleic acid sequences are administered into a host animal or human subject.
- polypeptide is preferably intended to refer to any amino acid chain length, including those of short peptides from about 2 to about 20 amino acid residues in length, oligopeptides from about 10 to about 100 amino acid residues in length, and longer polypeptides including from about 100 amino acid residues or more in length.
- polypeptides and proteins of the present invention also include polypeptides and proteins that are or have been post- translationally modified, and include any sugar or other derivative(s) or conjugate(s) added to the backbone amino acid chain.
- “Purified,” as used herein, means separated from many other compounds or entities.
- a compound or entity may be partially purified, substantially purified, or pure.
- a compound or entity is considered pure when it is removed from substantially all other compounds or entities, i.e., is preferably at least about 90%, more preferably at least about 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater than 99% pure.
- a partially or substantially purified compound or entity may be removed from at least 50%, at least 60%, at least 70%, or at least 80% of the material with which it is naturally found, e.g., cellular material such as cellular proteins and/or nucleic acids.
- the term “recombinant” indicates that the material (e.g, a polynucleotide or a polypeptide) has been artificially or synthetically (non-naturally) altered by human intervention. The alteration can be performed on the material within or removed from, its natural environment, or native state. Specifically, e.g, a promoter sequence is “recombinant” when it is produced by the expression of a nucleic acid segment engineered by the hand of man.
- a “recombinant nucleic acid” is one that is made by recombining nucleic acids, e.g, during cloning, DNA shuffling or other procedures, or by chemical or other mutagenesis
- a “recombinant polypeptide” or “recombinant protein” is a polypeptide or protein which is produced by expression of a recombinant nucleic acid
- a “recombinant virus,” e.g, a recombinant AAV virus is produced by the expression of a recombinant nucleic acid.
- regulatory element refers to a region or regions of a nucleic acid sequence that regulates transcription.
- exemplary regulatory elements include, but are not limited to, enhancers, post-transcriptional elements, transcriptional control sequences, and such like.
- RNA segment refers to an RNA molecule that has been isolated free of total cellular RNA of a particular species. Therefore, RNA segments can refer to one or more RNA segments (either of native or synthetic origin) that have been isolated away from, or purified free from, other RNAs. Included within the term “RNA segment,” are RNA segments and smaller fragments of such segments.
- sequence essentially as set forth in SEQ ID NO:X means that the sequence substantially corresponds to a portion of SEQ ID NO:X and has relatively few nucleotides (or amino acids in the case of polypeptide sequences) that are not identical to, or a biologically functional equivalent of, the nucleotides (or amino acids) of SEQ ID NO:X.
- biologically functional equivalent is well understood in the art, and is further defined in detail herein. Accordingly, sequences that have about 85% to about 90%; or more preferably, about 91% to about 95%; or even more preferably, about 96% to about 99%; of nucleotides that are identical or functionally equivalent to one or more of the nucleotide sequences provided herein are particularly contemplated to be useful in the practice of the invention.
- Suitable standard hybridization conditions for nucleic acids for use in the present invention include, for example, hybridization in 50% formamide, 5x Denhardt’s solution, 5x SSC, 25 mM sodium phosphate, 0.1% SDS and 100 pg/mL of denatured salmon sperm DNA at 42°C for 16 hr followed by 1 hr sequential washes with O.lx SSC, 0.1% SDS solution at 60°C to remove the desired amount of background signal.
- Lower stringency hybridization conditions for the present invention include, for example, hybridization in 35% formamide,
- 5x Denhardt’s solution 5x SSC, 25 mM sodium phosphate, 0.1% SDS and 100 pg/mL denatured salmon sperm DNA or A. coli DNA at 42°C for 16 hr followed by sequential washes with 0.8x SSC, 0.1% SDS at 55°C.
- structural gene is intended to generally describe a polynucleotide, such as a gene, that is expressed to produce an encoded peptide, polypeptide, protein, ribozyme, catalytic RNA molecule, or antisense molecule.
- subject describes an organism, including mammals such as primates, to which treatment with the compositions according to the present invention can be provided.
- Mammalian species that can benefit from the disclosed methods of treatment include, but are not limited to, apes; chimpanzees; orangutans; humans; monkeys; domesticated animals such as dogs and cats; livestock such as horses, cattle, pigs, sheep, goats, and chickens; and other animals such as mice, rats, guinea pigs, and hamsters.
- substantially complementary when used to define either amino acid or nucleic acid sequences, means that a particular subject sequence, for example, an oligonucleotide sequence, is substantially complementary to all or a portion of the selected sequence, and thus will specifically bind to a portion of an mRNA encoding the selected sequence.
- sequences will be highly complementary to the mRNA “target” sequence, and will have no more than about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, or about 10 or so base mismatches throughout the complementary portion of the sequence.
- sequences may be exact matches, i.e., be completely complementary to the sequence to which the oligonucleotide specifically binds, and therefore have zero mismatches along the complementary stretch.
- highly complementary sequences will typically bind quite specifically to the target sequence region of the mRNA and will therefore be highly efficient in reducing, and/or even inhibiting the translation of the target mRNA sequence into polypeptide product.
- Substantially complementary nucleic acid sequences will be greater than about 80 percent complementary (or “% exact-match”) to a corresponding nucleic acid target sequence to which the nucleic acid specifically binds, and will, more preferably be greater than about 85 percent complementary to the corresponding target sequence to which the nucleic acid specifically binds.
- nucleic acid sequences will be greater than about 90 percent complementary to the corresponding target sequence to which the nucleic acid specifically binds, and may in certain embodiments be greater than about 95 percent complementary to the corresponding target sequence to which the nucleic acid specifically binds, and even up to and including about 96%, about 97%, about 98%, about 99%, and even about 100% exact match complementary to all or a portion of the target sequence to which the designed nucleic acid specifically binds.
- Percent similarity or percent complementary of any of the disclosed nucleic acid sequences may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG).
- the GAP program utilizes the alignment method of Needleman and Wunsch (1970). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) that are similar, divided by the total number of symbols in the shorter of the two sequences.
- the preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess (1986), (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
- the term “substantially free” or “essentially free” in connection with the amount of a component preferably refers to a composition that contains less than about 10 weight percent, preferably less than about 5 weight percent, and more preferably less than about 1 weight percent of a compound. In preferred embodiments, these terms refer to less than about 0.5 weight percent, less than about 0.1 weight percent, or less than about 0.01 weight percent.
- structural gene is intended to generally describe a polynucleotide, such as a gene, that is expressed to produce an encoded peptide, polypeptide, protein, ribozyme, catalytic RNA molecule, or antisense molecule.
- subject describes an organism, including mammals such as primates, to which treatment with the compositions according to the present invention can be provided.
- Mammalian species that can benefit from the disclosed methods of treatment include, but are not limited to, humans, non-human primates such as apes; chimpanzees; monkeys, and orangutans, domesticated animals, including dogs and cats, as well as livestock such as horses, cattle, pigs, sheep, and goats, or other mammalian species including, without limitation, mice, rats, guinea pigs, rabbits, hamsters, and the like.
- nucleic acid or an amino acid sequence denote characteristics of a nucleic acid or an amino acid sequence, wherein a selected nucleic acid sequence or a selected amino acid sequence has at least about 70 or about 75 percent sequence identity as compared to a selected reference nucleic acid or amino acid sequence. More typically, the selected sequence and the reference sequence will have at least about 76, 77, 78, 79, 80, 81, 82, 83, 84 or even 85 percent sequence identity, and more preferably, at least about 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 percent sequence identity.
- sequences often share greater than at least about 96, 97, 98, or 99 percent sequence identity between the selected sequence and the reference sequence to which it was compared.
- synthetic shall mean that the material is not of a human or animal origin.
- Targeting moiety is any factor that may facilitate targeting of a specific site by a particle.
- the targeting moiety may be a chemical targeting moiety, a physical targeting moiety, a geometrical targeting moiety, or a combination thereof.
- the chemical targeting moiety may be a chemical group or molecule on a surface of the particle;
- the physical targeting moiety may be a specific physical property of the particle, such as a surface such or hydrophobicity;
- the geometrical targeting moiety includes a size and a shape of the particle.
- the chemical targeting moiety may be a dendrimer, an antibody, an aptamer, which may be a thioaptamer, a ligand, an antibody, or a biomolecule that binds a particular receptor on the targeted site.
- a physical targeting moiety may be a surface charge.
- the charge may be introduced during the fabrication of the particle by using a chemical treatment such as a specific wash. For example, immersion of porous silica or oxidized silicon surface into water may lead to an acquisition of a negative charge on the surface.
- the surface charge may be also provided by an additional layer or by chemical chains, such as polymer chains, on the surface of the particle.
- polyethylene glycol chains may be a source of a negative charge on the surface.
- Polyethylene glycol chains may be coated or covalently coupled to the surface using methods known to those of ordinary skill in the art.
- therapeutically-practical period means the period of time that is necessary for one or more active agents to be therapeutically effective.
- therapeutically-effective refers to reduction in severity and/or frequency of one or more symptoms, elimination of one or more symptoms and/or underlying cause, prevention of the occurrence of symptoms and/or their underlying cause, and the improvement or a remediation of damage.
- a “therapeutic agent” may be any physiologically or pharmacologically active substance that may produce a desired biological effect in a targeted site in a subject.
- the therapeutic agent may be a chemotherapeutic agent, an immunosuppressive agent, a cytokine, a cytotoxic agent, a nucleolytic compound, a radioactive isotope, a receptor, and a pro-drug activating enzyme, which may be naturally occurring, produced by synthetic or recombinant methods, or a combination thereof.
- Drugs that are affected by classical multidrug resistance such as vinca alkaloids (e.g., vinblastine and vincristine), the anthracyclines (e.g, doxorubicin and daunorubicin), RNA transcription inhibitors (e.g, actinomycin-D) and microtubule stabilizing drugs (e.g, paclitaxel) may have particular utility as the therapeutic agent.
- Cytokines may be also used as the therapeutic agent. Examples of such cytokines are lymphokines, monokines, and traditional polypeptide hormones.
- An anti- neurodegenerative agent may be a preferred therapeutic agent. For a more detailed description of such agents and other therapeutic agents, those skilled in the art are referred to any number of instructive manuals including, but not limited to, the Physician ’s Desk Reference and Hardman and Limbird (2001).
- transcription factor recognition site and a “transcription factor binding site” refer to a polynucleotide sequence(s) or sequence motif(s), which are identified as being sites for the sequence-specific interaction of one or more transcription factors, frequently taking the form of direct protein-DNA binding.
- transcription factor binding sites can be identified by DNA footprinting, gel mobility shift assays, and the like, and/or can be predicted based on known consensus sequence motifs, or by other methods known to those of ordinary skill in the art.
- Transcriptional regulatory element refers to a polynucleotide sequence that activates transcription alone or in combination with one or more other nucleic acid sequences.
- a transcriptional regulatory element can, for example, comprise one or more promoters, one or more response elements, one or more negative regulatory elements, and/or one or more enhancers.
- Transcriptional unit refers to a polynucleotide sequence that comprises at least a first structural gene operably linked to at least a first cA-acting promoter sequence and optionally linked operably to one or more other cA-acting nucleic acid sequences necessary for efficient transcription of the structural gene sequences, and at least a first distal regulatory element as may be required for the appropriate tissue-specific and developmental transcription of the structural gene sequence operably positioned under the control of the promoter and/or enhancer elements, as well as any additional cA- sequences that are necessary for efficient transcription and translation (e.g., polyadenylation site(s), mRNA stability controlling sequence(s), etc.
- the term “transformation” is intended to generally describe a process of introducing an exogenous polynucleotide sequence (e.g., a viral vector, a plasmid, or a recombinant DNA or RNA molecule) into a host cell or protoplast in which the exogenous polynucleotide is incorporated into at least a first chromosome or is capable of autonomous replication within the transformed host cell.
- Transfection, electroporation, and “naked” nucleic acid uptake all represent examples of techniques used to transform a host cell with one or more polynucleotides.
- the term “transformed cell” is intended to mean a host cell whose nucleic acid complement has been altered by the introduction of one or more exogenous polynucleotides into that cell.
- Treating refers to providing any type of medical or surgical management to a subject. Treating can include, but is not limited to, administering a composition comprising a therapeutic agent to a subject. “Treating” includes any administration or application of a compound or composition of the invention to a subject for purposes such as curing, reversing, alleviating, reducing the severity of, inhibiting the progression of, or reducing the likelihood of a disease, disorder, or condition or one or more symptoms or manifestations of a disease, disorder, or condition.
- compositions of the present invention may also be administered prophylactically, i.e., before development of any symptom or manifestation of the condition, where such prophylaxis is warranted.
- the subject will be one that has been diagnosed for being “at risk” of developing such a disease or disorder, either as a result of familial history, medical record, or the completion of one or more diagnostic or prognostic tests indicative of a propensity for subsequently developing such a disease or disorder.
- vector refers to a nucleic acid molecule (typically comprised of DNA) capable of replication in a host cell and/or to which another nucleic acid segment can be operatively linked so as to bring about replication of the attached segment.
- a plasmid, cosmid, or a virus is an exemplary vector.
- nucleic acid segments of the present invention in combination with an appropriate detectable marker (i.e., a “label,”), such as in the case of employing labeled polynucleotide probes in determining the presence of a given target sequence in a hybridization assay.
- an appropriate detectable marker i.e., a “label,”
- a wide variety of appropriate indicator compounds and compositions are known in the art for labeling oligonucleotide probes, including, without limitation, fluorescent, radioactive, enzymatic or other ligands, such as avi din/biotin, etc., which are capable of being detected in a suitable assay.
- an enzyme tag such as urease, alkaline phosphatase or peroxidase
- colorimetric, chromogenic, or fluorogenic indicator substrates are known that can be employed to provide a method for detecting the sample that is visible to the human eye, or by analytical methods such as scintigraphy, fluorimetry, spectrophotometry, and the like, to identify specific hybridization with samples containing one or more complementary or substantially complementary nucleic acid sequences.
- multiplexing assays where two or more labeled probes are detected either simultaneously or sequentially, it may be desirable to label a first oligonucleotide probe with a first label having a first detection property or parameter (for example, an emission and/or excitation spectral maximum), which also labeled a second oligonucleotide probe with a second label having a second detection property or parameter that is different (i.e., discreet or discernible from the first label.
- first detection property or parameter for example, an emission and/or excitation spectral maximum
- the method involves the growth and manipulation of patient Tregs outside of the body.
- the methods for the production of therapeutic populations of Tregs provided herein may be useful for treating patients with a pathological disease or condition. Also provided herein are therapeutic populations of Tregs produced by methods described herein and pharmaceutical compositions thereof.
- the therapeutic populations of Tregs produced by a method provided herein have advantageous properties for clinical application.
- a therapeutic population of Tregs produced by a method provided herein may be cryopreserved without loss of viability, purity or potency.
- a therapeutic population of ex v/vo-expanded Tregs produced by a method provided herein may be cryopreserved, thawed and without further expansion, demonstrate maintenance of viability, purity and potency as compared to the expanded Tregs prior to cryopreservation.
- a therapeutic population of Tregs produced by a method described herein comprises Tregs with higher suppressive ability than the Tregs enriched from the donor samples, or compared to a healthy donor’s Tregs.
- a therapeutic population of Tregs produced by a method described herein comprises Tregs with a suppressive ability that is absent Tregs enriched from the donor samples, or compared to a healthy donor’s Tregs.
- a method of producing a therapeutic population of Tregs provided herein is an improved method compared to methods known in the art.
- the method of producing a therapeutic population of Tregs comprises the steps of (1) enriching a cell population obtained from a subject for Tregs; (2) ex vivo expansion of the cell population enriched for Tregs and/or (3) cryopreservation of the expanded Tregs.
- a population of cells comprising Tregs may be enriched from a biological sample, e.g., a peripheral blood sample or thymic tissue.
- methods of producing a therapeutic population of Tregs provided herein comprise a step of enriching Tregs from in a biological donor sample, e.g., a peripheral blood sample or thymic tissue.
- the therapeutic population of Tregs is obtained from a serum sample suspected of containing Tregs.
- the therapeutic population of Tregs is obtained from a cell sample suspected of containing Tregs, obtained from a donor via leukapheresis.
- the therapeutic population of Tregs is obtained from a biological sample suspected of containing Tregs.
- the therapeutic population of Tregs is enriched from a biological sample from a donor subject, in particular a human donor subject.
- the biological sample can be any sample suspected of containing Tregs, likely to contain Tregs or know to contain Tregs.
- Such biological samples may be taken directly from the subject, or may be samples resulting from one or more processing steps, such as separation, e.g. selection or enrichment, centrifugation, washing, and/or incubation.
- Biological samples include, but are not limited to, body fluids, such as blood, plasma, serum, cerebrospinal fluid, synovial fluid, tissue and organ samples, including processed samples derived therefrom.
- the sample is blood or a blood-derived sample, or is or is derived from an apheresis or leukapheresis product.
- exemplary samples include whole blood, peripheral blood mononuclear cells (PBMCs), leukocytes, bone marrow, and thymus.
- the biological sample is a blood-derived sample, e.g., a samples derived from whole blood, serum, or plasma.
- the biological sample is or includes peripheral blood mononuclear cells.
- the biological sample is a peripheral blood or serum sample.
- the biological sample is a lymph node sample.
- the donor subject is a human subject. In some embodiments, the human donor is a healthy donor.
- the donor subject is diagnosed with or is suspected of having a disorder associated with Treg dysfunction. In some embodiments, the donor subject is diagnosed with or is suspected of having a disorder associated with Treg deficiency. In some embodiments, the donor subject is diagnosed with or is suspected of having a condition driven by a T cell response.
- the donor subject is diagnosed with or is suspected of having a neurodegenerative disease. In some embodiments, the donor subject is diagnosed with or is suspected of having Alzheimer’s disease, Amyotrophic Lateral Sclerosis, Huntington’s disease or frontotemporal dementia.
- the donor subject is diagnosed with or is suspected of having a disorder that would benefit from downregulation of the immune system.
- the donor subject is diagnosed with or suspected of having an autoimmune disease.
- the autoimmune disease may be, for example, systemic sclerosis (scleroderma), polymyositis, ulcerative colitis, inflammatory bowel disease, Crohn’s disease, celiac disease, multiple sclerosis (MS), rheumatoid arthritis (RA), Type I diabetes, psoriasis, dermatomyositis, systemic lupus erythematosus, cutaneous lupus, myasthenia gravis, autoimmune nephropathy, autoimmune hemolytic anemia, autoimmune cytopenia autoimmune hepatitis, autoimmune uveitis, alopecia, thyroiditis or pemhigus.
- the donor subject is diagnosed with or suspected of having heart failure or ischemic cardiomyopathy.
- the donor subject is diagnosed with or suspected of having graft-versus-host disease, e.g., after undergoing organ transplantation (such as a kidney transplantation or a liver transplantation), or after undergoing stem cell transplantation (such as hematopoietic stem cell transplantation).
- organ transplantation such as a kidney transplantation or a liver transplantation
- stem cell transplantation such as hematopoietic stem cell transplantation.
- the donor subject is diagnosed with or suspected of having neuroinflammation.
- Neuroinflammation may be associated, for example, with stroke, acute disseminated encephalitis, acute optic neuritis, transverse myelitis, neuromyelitis optica, epilepsy, traumatic brain injury, spinal cord injury, encephalitis central nervous system (CNS) vasculitis, neurosarcoidosis, autoimmune or post-infectious encephalitis or chronic meningitis.
- the donor subject is diagnosed with or suspected of having chronic inflammatory demyelinating polyradiculoneuropathy (CIDP).
- CIDP chronic inflammatory demyelinating polyneuropathy
- AIDP acute inflammatory demyelinating polyneuropathy
- the donor subject is diagnosed with or suspected of having Guillain-Barre syndrome (GBS).
- the donor subject is diagnosed with or suspected of having cardo-inflammation, e.g., cardio-inflammation associated with myocardial infarction, ischemic cardiomyopathy, with heart failure.
- cardo-inflammation e.g., cardio-inflammation associated with myocardial infarction, ischemic cardiomyopathy, with heart failure.
- the donor subject has had a stroke.
- the donor subject is diagnosed with or suspected of having cancer, e.g., a blood cancer.
- the donor subject is diagnosed with or suspected of having asthma.
- the donor subject is diagnosed with or suspected of having eczema.
- the donor subject is diagnosed with or suspected of having a disorder associated with overactivation of the immune system.
- the donor subject is diagnosed with or suspected of having Tregopathy.
- the Tregopathy may be caused by a FOXP3, CD25, cytotoxic T lymphocyte- associated antigen 4 (CTLA4), LPS-responsive and beige-like anchor protein (LRBA), or BTB domain and CNC homolog 2 (BACH2) gene loss-of-function mutation, or a signal transducer and activator of transcription 3 (STAT3) gain-of-function mutation.
- CTL4 cytotoxic T lymphocyte- associated antigen 4
- LRBA LPS-responsive and beige-like anchor protein
- BACH2 BTB domain and CNC homolog 2
- STAT3 signal transducer and activator of transcription 3
- lymphocytes may be obtained from a peripheral blood sample by leukapheresis.
- Tregs are enriched from a population of lymphocytes.
- repeated peripheral blood samples are obtained from a donor for producing Tregs.
- two or more peripheral blood samples are obtained from a donor.
- insufficient Tregs are obtained from a donor sample after expanding for 25 days and a subsequent sample is obtained.
- the donor sample undergoes volume reduction (e.g., volume reduction by a method described herein, such as a method described in Section 8.5.3) during the enrichment process.
- biological samples e.g., leukapheresis samples
- biological samples from more than one donor are pooled prior to the enrichment process to generate an allogeneic population of Tregs.
- biological samples e.g., leukapheresis samples from 2, 3, 4, or 5 donors are pooled.
- Tregs may be enriched from a biological sample by any method known in the art or described herein (e.g., a method described in section 8).
- Tregs are enriched from a sample using magnetic bead separation (e.g., CliniMACS Tubing Set LS (162-01) or CliniMACS® Plus Instrument), fluorescent cell sorting, and disposable closed cartridge based cell sorters.
- Enrichment for cells expressing one or more markers refers to increasing the number or percentage of such cells in the population of cells, but does not necessarily result in a complete absence of cells not expressing the marker. Depletion of cells expressing one or more markers refers to decreasing the number or percentage of such cells in the population of cells, but does not necessarily result in a complete removal of all cells expressing such marker or markers.
- the enrichment comprises a step of affinity- or immunoaffmity-based separation of cells expressing one or more markers (e.g., Treg cell surface markers).
- markers e.g., Treg cell surface markers.
- Such separation steps can be based on positive selection, in which the cells expressing one or more markers are retained, and/or on negative selection (depletion), in which the cells not expressing one or more markers are retained.
- the separation may be based on the expression (e.g., positive or negative expression) or expression level (e.g., high or low expression) of one or more markers (e.g., Treg cell surface markers).
- “high expression” and “low expression” are generally relative to the whole population of cells.
- separation of cells may be based on CD8 expression.
- separation of cells may be based on CD 19 expression.
- separation of cells may be based on high CD25 expression.
- enrichment of Tregs may comprise incubation with an antibody or binding partner that specifically binds to a marker (e.g., a Treg cell surface marker), followed generally by washing steps and separation of cells having bound the antibody or binding partner from those cells having not bound to the antibody or binding partner.
- a marker e.g., a Treg cell surface marker
- the antibody or binding partner is bound to a solid support or matrix, such as a sphere or bead, for example a nanoparticle, microbeads, nanobeads, including agarose, magnetic bead or paramagnetic beads.
- a solid support or matrix such as a sphere or bead, for example a nanoparticle, microbeads, nanobeads, including agarose, magnetic bead or paramagnetic beads.
- the spheres or beads can be packed into a column to effect immunoaffmity chromatography.
- the antibody or binding partner is detectably labeled.
- the antibody or binding partner is attached to small, magnetically responsive particles or microparticles, such as nanoparticles or paramagnetic beads.
- Such beads are known and are commercially available (e.g., Dynabeads® (Life Technologies, Carlsbad, CA), MACS® beads (Miltenyi Biotec, San Diego, CA) or Streptamer® bead reagents (IB A, Germany)).
- Such particles or microparticles may be incubated with the population of cells to be enriched and then placed in a magnetic field. This results in those cells that are attached to the particles or microparticles via the antibody or binding partner being attracted to the magnet and separated from the unbound cells. This method allows for retention of the cells attached to the magnet (positive selection) or removal of the cells attracted to the magnet (negative selection).
- a method of producing a therapeutic population of Tregs provided herein comprises both positive and negative selection during the enrichment step.
- the biological sample is obtained within about 25-35min, about 35-45min, about 45-60min, about 60-75 min, about 75-90min, about 90-120min, about 120-150min, about 150-180min, about 2-3h, about 3-4h, about 4-5h or about 5-6h of the beginning of the enriching step.
- the sample is obtained within about 30min of the beginning of the enriching step.
- the biological sample is not stored (e.g., stored at 4°C) over night.
- enrichment of Tregs from a human sample comprises depleting the sample of CD8+ cells. In some embodiments, enrichment of Tregs from a human sample comprises depleting a sample of CD 19+ cells. In some embodiments, enrichment of Tregs from a biological sample comprises depleting the sample of CD8+ cells and CD19+ cells. In some embodiments, enrichment of Tregs from a biological sample comprises enriching the cell population for CD25high cells. In some embodiments, enrichment of Tregs from a biological sample comprises depletion of CD8+ cells and CD19+ cells from the sample and enriching the cell population for CD25high cells.
- the population of cells enriched for Tregs comprises an increased proportion of CD4+CD25high Tregs relative to the proportion of CD4+CD25high Tregs in the Tregs prior to enrichment as determined by flow cytometry.
- the proportion of CD4+CD25high Tregs is increased by about 2-fold to about 4-fold, about 4-fold to about 6-fold, about 6-fold to about 8-fold, about 8-fold to about 10- fold, about 10-fold to about 15-fold, about 15-fold to about 20-fold, about 20-fold to about 25-fold, about 25-fold to about 30-fold, about 30-fold to about 35-fold, about 35-fold to about 40-fold, about 40-fold to about 45-fold, about 45-fold to about 50-fold.
- the population of cells enriched for Tregs comprises an increased proportion of CD4+CD25 high CD127 low Tregs relative to the proportion of CD4+CD25highCD1271ow Tregs in the Tregs prior to enrichment as determined by flow cytometry.
- the proportion of CD4+CD25 high CD127 low Tregs is increased by about 2-fold to about 4-fold, about 4-fold to about 6-fold, about 6-fold to about 8-fold, about 8-fold to about 10-fold, about 10-fold to about 15-fold, about 15-fold to about 20-fold, about 20-fold to about 25-fold, about 25-fold to about 30-fold, about 30-fold to about 35-fold, about 35-fold to about 40-fold, about 40-fold to about 45-fold, about 45-fold to about 50-fold.
- the population of cells enriched for Tregs comprises CD25+ Tregs wherein the expression of CD25 in the Tregs is increased relative to the expression of CD25 in the Tregs prior to enrichment, as determined by flow cytometry.
- the expression of CD25 is increased by at least about 5-fold, at least about 10-fold, at least about 15-fold, at least about 20-fold, at least about 25-fold, at least about 30-fold, at least about 35-fold, at least about 40-fold, at least about 45-fold, or at least about 50-fold.
- the population of cells enriched for Tregs comprises CD127+ Tregs wherein the expression of CD127 in the Tregs is increased relative to the expression of CD127 in the Tregs prior to enrichment, as determined by flow cytometry.
- the expression of CD127 is increased by at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, or at least about 3-fold.
- the granularity of the Tregs in the enriched population of Tregs is increased relative to the granularity of the Tregs prior to enrichment, as determined by flow cytometry. In specific embodiments, the granularity of the Tregs increased by at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, or at least about 3-fold. [00216] In some embodiments, the size of the Tregs in the enriched population of Tregs is increased relative to the size of the Tregs prior to enrichment, as determined by flow cytometry. In specific embodiments, the size of the Tregs increased by at least about 1.2- fold, at least about 1.5-fold, or at least about 2-fold. 6.1.2. Methods of Expanding Tregs Ex Vivo
- a method provided herein comprises a step of expanding the therapeutic population of Tregs enriched from a biological sample.
- This expansion of the therapeutic population of Tregs may comprise culturing the cells that have been enriched from a biological samples in media, for example, in serum-free media (e.g., TexMACS Medium).
- the cells enriched from a biological sample are cultured about 37°C and about 5% CO2.
- the cells enriched from a biological sample are cultured out under good manufacturing practice (GMP) conditions.
- the cells enriched from a biological sample are cultured in a closed system.
- the expansion of the therapeutic population of Tregs begins within 25-35 min, within 20-40 min, within 15-45 min or within 10-50 min of the enrichment from a biological sample. In some embodiments, the expansion of the therapeutic population of Tregs begins within about 30min of the enrichment from a biological sample.
- Tregs may be expanded ex vivo by culturing the cells in the presence of one or more expansion agents.
- the expansion agent is IL-2.
- the appropriate concentration of IL-2 in the culture media can be determined by a person of skill in the art.
- the concentration of IL-2 in the cell culture media is about 5-10 IU/mL, about 10-20 IU/mL, about 20-30 IU/mL, about 30-40 IU/mL, about 40-50 IU/mL, about 50-100 IU/mL, about 100-200 IU/mL, about 200-300 IU/mL, about 300-400 IU/mL, about 400-500 IU/mL, about 500-600 IU/mL, about 600-700 IU/mL, about 700-800 IU/mL, about 800-900 IU/mL, about 900-1000 IU/mL, about 1000-1500 IU/mL, about 1500-2000 IU/mL, about 2000-2500 IU/mL, about 2500-3000 IU/mL, about 3000-3500 IU/mL, about 3500-4000 IU/mL, about 4000-4500 IU/mL, about 4500-5000 IU/mL, about 5000-6000 IU/
- the expansion agent activates CD3, e.g., the expansion agent is an anti-CD3 antibody.
- the expansion agent activates CD28, e.g., the expansion agent is an anti-CD28 antibody.
- the expansion agent is a soluble anti-CD3 antibody.
- the anti-CD3 antibody is OKT3.
- the concentration of soluble anti-CD3 antibody in the culture media is about 0.1-0.2 ng/mL, about 0.2-0.3 ng/mL, about 0.3-0.4 ng/mL, about 0.4-0.5 ng/mL about 0.5-1 ng/mL, about 1- 5 ng/mL, about 5-10 ng/mL, about 10-15 ng/mL, about 15-20 ng/mL, about 20-25 ng/mL, about 25-30 ng/mL, about 30-35 ng/mL, about 35-40 ng/mL, about 40-45 ng/mL, about 45- 50 ng/mL, about 50-60 ng/mL, about 60-70 ng/mL, about 70-80 ng/mL, about 80-90 ng/mL, or about 90-100 ng/mL.
- the expansion agent is a soluble anti-CD28 antibody.
- anti-CD28 antibodies include NA/LE (e.g. BD Pharmingen), IM1376 (e.g. Beckman Coulter), or 15E8 (e.g. Miltenyi Biotec).
- the concentration of soluble anti-CD28 antibody in the culture media is about 1-2 ng/mL, about 2-3 ng/mL, about 3-4 ng/mL, about 4-5 ng/mL, about 5-10 ng/mL, about 10-15 ng/mL, about 15-20 ng/mL, about 20-25 ng/mL, about 25-30 ng/mL, about 30-35 ng/mL, about 35-40 ng/mL, about 40-45 ng/mL, about 45-50 ng/mL, about 50-60 ng/mL, about 60-70 ng/mL, about 70-80 ng/mL, about 80-90 ng/mL, about 90-100 ng/mL, about 100-200 ng/mL, about 200-300 ng/mL, about 300-400 ng/mL, about 400-500 ng/mL, 500-600 ng/mL, 600-700 ng/mL, about 700-800 ng/mL, about 800-900
- both an anti-CD3 antibody and an anti-CD28 antibody are present in the cell culture media.
- the anti-CD3 antibody and the anti- CD28 antibody are attached to a solid surface.
- the anti-CD3 antibody and the anti-CD28 antibody are attached to beads.
- beads e.g., 3.5 pm particles loaded with CD28 antibodies, anti -biotin antibodies and CD3 -Biotin are present in the cell culture medium Such beads are commercially available (e.g., MACS GMP ExpAct Treg Kit, DYNABEADS ® M-450 CD3/CD28 T Cell Expander).
- the ratio of anti-CD3 antibody to anti-CD28 antibody on the beads is about 100:1, 90:1, 80:1, 70:1, 60:1, 50:1, 40:1, 30:1, 20:1, 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:20, 1:30, 1:40, 1:50, 1:60, 1:70, 1:80, 1:90 or 1 : 100.
- the population of Tregs is cultured in the presence of both IL-2 and beads loaded with CD28 antibodies, anti -biotin antibodies and CD3 -Biotin.
- the beads coated with anti-CD3 and anti-CD28 antibody are first added to the culture within about 4-5 days of initiating culture. In some embodiments, the beads coated with anti-CD3 and anti-CD28 antibody are again added to the culture medium about 14 days after the beads coated with anti-CD3 and anti-CD28 antibody were first added to the culture medium. In specific embodiments, the ratio of beads to cells in the culture is 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1 or 1:1.
- the expansion agent or agents may be added to the culture medium every 1, 2, 3, 4, or 5 days. In specific embodiments, the expansion agent is added to the culture medium every 2-3 days. In other specific embodiments, the expansion agent is added to the culture medium on day 6, 8, and 11, wherein day 0 is the day on which the biological sample is obtained from the subject. In some specific embodiments, the expansion agent is not added to the culture medium on day 13, wherein day 0 is the day on which the biological sample is obtained from the subject.
- the one or more expansion agent is first added to the culture within about 16-18 hours, within 18-24h, within 24-36h, within 36-48h, within about 24h, within about 24h, within about 3 days, within about 4 days, within about 5 days, within about 6 days, or within about 7 days of initiating culture. In some embodiments, the one or more expansion agent is first added to the culture within about 4-5 days of initiating culture. In some embodiments, the one or more expansion agent is again added to the culture medium about 14 days after the expansion agent was first added to the culture medium.
- no expansion agent is added to the culture on a given day, that day is considered a “rest day.” In some embodiments, no expansion agent is administered during the day preceding the day on which the therapeutic population of Tregs is harvested. In some embodiments, no expansion agent is administered during the 2 days, 3 days, 4 days, 5 days or 6 days preceding the day on which the therapeutic population of Tregs is harvested.
- the therapeutic population of Tregs may be expanded ex vivo by culturing the cells in the presence of one or more agents that inhibit mammalian target of rapamycin (mTor).
- the mTor inhibitor is rapamycin.
- the mTor inhibitor is an analog of rapamycin (a “rapalog,” e.g., Temsirolimus, Everolimus, or Ridaforolimus).
- the mTor inhibitor is ICSN3250, OSU-53, or AZD8055.
- the concentration of rapamycin in the cell culture medium is about 1-20 nmol/L, about 20-30 nmol/L, about 30-40 nmol/L, about 40-50 nmol/L, about 50-60 nmol/L, about 60-70 nmol/L, about 70-80nmol/L, about 80- 90 nmol/L, about 90-100 nmol/L, about 100-150 nmol/L, about 150-200 nmol/L, about 200- 250 nmol/L, about 250-300 nmol/L, about 300-350 nmol/L, about 350-400 nmol/L, about 400-450nmol/L, about 450-500 nmol/L, about 500-600 nmol/L, about 600-700 nmol/L, about 700-800 nmol/L, about 800-900 nmo/L or about 900-1000 nmol/L. In some embodiments, the concentration of rapamycin in the cell culture media is about 100 nmol/L,
- the mTor inhibitor is first added to the culture within about 16-18 hours, within 18-24h, within 24-36h, within 36-48h, within about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days of initiating culture. In some embodiments, the mTor inhibitor is added to the culture medium about every 2-3 days.
- the therapeutic population of Tregs may be expanded by culturing them for an appropriate duration of time. The time required for expansion resulting in a sufficiently expanded therapeutic population of Tregs for therapeutic application may be readily determined by a person of skill in the art, e.g., by monitoring the proportion of CD4+CD25+cells using flow cytometry.
- a sufficiently expanded therapeutic population of Tregs is a population of cells that contains more than 70% CD4+CD25+ cells as determined by flow cytometry.
- a sufficiently expanded therapeutic population of Tregs is a population that contains about 1 x 10 6 to about 2 x 10 6 , about 2 x 10 6 to about 3 x 10 6 , about 3 x 10 6 to about 4 x 10 6 , about 4 x 10 6 to about 5 x 10 6 , about 5 x 10 6 to about 6 x 10 6 , about 6 x 10 6 to about 7 x 10 6 , about 7 x 10 6 to about 8 x 10 6 , about 8 x 10 6 to about 9 x 10 6 , about 9 x 10 6 to about 1 x 10 7 , about 1 x 10 7 to about 2 x 10 7 , about 2 x 10 7 to about 3 x 10 7 , about 3 x 10 7 to about 4 x 10 7 , about 4 x 10 7
- a sufficiently expanded therapeutic population of Tregs is a population that contains lxlO 6 CD4+CD25+cells (+/- 10%) per kg of body weight of the intended recipient of the therapeutic population of Tregs as determined by flow cytometry.
- a sufficiently expanded therapeutic population of Tregs is a population of cells that contains about or more than about 70% CD4+CD25+ cells and contains lxlO 6 CD4+CD25+cells (+/- 10%) per kg of body weight of the intended recipient of the therapeutic population of Tregs as determined by flow cytometry.
- the intended recipient of the therapeutic population of Tregs may be the same subject as the donor of the biological sample from which the Tregs were enriched.
- the number of CD4+CD25+cells may be determined every day, or every 2, 3, 4, or 5 days. If the culture does not contain a sufficiently expanded therapeutic population of Tregs on Day 15 (wherein day 0 is the day on which the biological sample is obtained from the subject), the cells may be re-activated with one or more expansion agents. If the culture does contain a sufficiently expanded therapeutic population of Tregs on Day 15 (wherein day 0 is the day on which the biological sample is obtained from the subject), the cells may harvested.
- a therapeutic population of Tregs is expanded by culturing for about 6-30 days, about 10-30 days, about 15-25 days, or about 18-22 days. In some embodiments, a therapeutic population of Tregs is expanded by culturing for about 15, 16,
- a therapeutic populatin of Tregs is expanded by culturing for about 6-15 days, about 8-15, about 8-12 days, or about 6, 7, 8, 9, 10, 11 or 12 days.
- Viability of the cells being expanded in culture may be determined using any method known in the art.
- the viability of cells being expanded in culture may be determined using trypan blue exclusion.
- Trypan blue is a dye which is excluded by cells with an intact membrane (viable cells) but taken up by cells with compromised membrane integrity (non-viable cells). Thus, viable cells appear clear under a light microscope, whereas non-viable cells appear blue. Equal amounts of trypan blue and cell suspension are mixed and counted. Viability is expressed as a percentage of trypan blue excluding cells.
- a therapeutic population of Tregs comprises about 60%, 65% or 70% viable cells as determined by trypan blue exclusion.
- a therapeutic population of Tregs comprises more than about 70% viable cells as determined by trypan blue exclusion.
- a therapeutic population of Tregs comprises about 75%, 80%, 85%, 90%, 95% or greater that 95% viable cells as determined by trypan blue exclusion.
- viability of the cells being expanded in culture is determined every 2-3 days. In some embodiments, viability of the cells being expanded in culture is determined every day or every 2, 3, 4, or 5 days.
- the cells are washed one or more times during the culturing to remove agents present during the incubation or culturing and/or to replenish the culture medium with one or more additional agents.
- the cells are washed during the incubation or culturing to reduce or remove the expansion agent(s).
- the culture medium may be replaced about every 2, 3, 4, 5, 6 or 7 days, for example, every 2-3 days. In some embodiments, only part of the culture medium (e.g., about 50% of the culture medium) is replaced. In other embodiments, the entire culture medium is replaced.
- the cell culture is not centrifuged during a change of culture medium. In some embodiments, the cell culture is not centrifuged during harvesting.
- the therapeutic population of Tregs may be harvested by any means known in the art, for example, by centrifugation.
- the therapeutic population of Tregs is harvested on day 15, wherein day 0 is the day on which the biological sample is obtained from the subject.
- the therapeutic population of Tregs is harvested on day 19, wherein day 0 is the day on which the biological sample is obtained from the subject.
- the therapeutic population of Tregs is harvested on day 20, wherein day 0 is the day on which the biological sample is obtained from the subject.
- the therapeutic population of Tregs is harvested on day 25, wherein day 0 is the day on which the biological sample is obtained from the subject.
- the therapeutic population of Tregs is harvested on day 16, 17 or 18, wherein day 0 is the day on which the biological sample is obtained from the subject. In some embodiments, the therapeutic population of Tregs is harvested on day 21, 22, 23, or 24, wherein day 0 is the day on which the biological sample is obtained from the subject.
- the population of Tregs is subjected to genetic engineering at any point of the method prior to cryopreservation.
- the population of Tregs is subjected to genetic engineering two or more times at any point prior to cryopreservation.
- the engineering may comprise the introduction of a transgene into the Tregs or the introduction of an mRNA into the Tregs.
- the genetic engineering may also comprise the gene editing through CRISPR-Cas9.
- the genetic engineering comprises the reduction of gene expression through siRNA or antisense oligonucleotides.
- the genetic engineering may allow for the use of a therapeutic population of Tregs provided herein in an allogeneic setting.
- the genetic engineering introduces a chimeric antigen receptor (CAR) into the Tregs.
- CAR chimeric antigen receptor
- cryopreserved population of Tregs having characteristics as described herein.
- a cryopreserved therapeutic population of Tregs may be produced by the methods described herein.
- pharmaceutical compositions comprising a cryopreserved therapeutic population of Tregs that has been thawed and is present in a formulation suitable for administration to a subject, for example a human subject.
- the formulation comprises a pharmaceutically acceptable carrier, e.g., normal saline.
- the formulation comprises human serum albumin.
- the formulation comprises normal saline and human serum albumin.
- a therapeutic population of Tregs is cryopreserved after expansion.
- the therapeutic population of Tregs may be cryopreserved in any suitable medium known in the art. Examples of media suitable for cryopreservation include, e.g., CryoStor ® CS10.
- a therapeutic population of Tregs is frozen in a composition comprising a cryoprotectant , for example, in a composition comprising DMSO (e.g., 10% DMSO).
- the therapeutic population of Tregs is frozen in a comprising glycerol.
- the cryoprotectant is or comprises DMSO and/or glycerol.
- a therapeutic population of Tregs may be stored at about - 200 °C to -190 °C, about -180 to -140 °C, or about -90 to -70 °C. In some embodiments, a therapeutic population of Tregs may be stored at about -196 °C. In some embodiments, a therapeutic population of Tregs may be stored at about -80 °C. In some embodiments, a therapeutic population of Tregs may be stored in the liquid nitrogen vapor phase. In some embodiments, a therapeutic population of Tregs may be stored on frozen carbon dioxide (dry ice).
- a cryopreserved therapeutic population of Tregs may be stored at a first temperature for a period of time, for example an extended period of time, e.g., about month, about 3 months, about 6 months, about 9 months, about 12 months, about 18 months, or about 24 months, and subsequently stored at a second temperature for a shorter period of time (e.g., about 6 hours, about 12 hours, about 24 hours, about 36 hours, or about 48 hours).
- a cryopreserved therapeutic population of Tregs may be stored at a first temperature for a period of time, e.g., about 6 hours, about 12 hours, about 24 hours, about 36 hours, or about 48 hours, subsequently stored at a second temperature for a longer period of time e.g., about month, about 3 months, about 6 months, about 9 months, about 12 months, about 18 months, or about 24 months.
- the first temperature is lower than the second temperature.
- the first temperature is about -200 °C to -190 °C, about -180 to -140 °C, about -90 to -70 °C, about -196 °C or about -80 °C.
- the second temperature is about -80 °C or about -20 °C.
- a therapeutic population of Tregs is stored at about -196 °C for about 1 month, about 3 months, about 6 months, about 9 months, about 12 months, about 18 months, or about 24 months and subsequently stored on frozen carbon dioxide for about 6 hours, about 12 hours, about 24 hours, about 36 hours, or about 48 hours.
- a therapeutic population of Tregs is stored in the liquid nitrogen vapor phasefor about 1 month, about 3 months, about 6 months, about 9 months, about 12 months, about 18 months, or about 24 months and subsequently stored on frozen carbon dioxide for about 6 hours, about 12 hours, about 24 hours, about 36 hours, or about 48 hours.
- the therapeutic population of Tregs is cryopreserved at a high Treg density.
- the therapeutic population of Tregs is cryopreserved at a concentration of lxlO 6 cells (+/- 10%) per kg of body weight of the intended recipient of the Tregs.
- the intended recipient of the Tregs may be the same or different individual as the subject from whom the initial biological sample containing Tregs was obtained (the donor).
- the cryopreserved therapeutic population of Tregs is stored at a density of at 10 million, at least 20 million, at least 25 million, at least 30 million, at least 35 million, at least 40 million, at least 45 million, at least 50 million, at least 55 million, at least 60 million, at least 65 million, at least 70 million, at least 75 million, at least 80 million, at least 85 million, at least 90 million, at least 95 million, or at least 100 million cells per mL.
- the therapeutic population of Tregs is cryopreserved in a cryovial.
- the therapeutic population of Tregs is frozen in a cryovial in a volume of about 0.5 mL to lmL, about 1 mL to 1.5 mL, or about 1.5 mL to 2 mL, about 2-5 mL, about 5-10 mL, or about 15-20 mL.
- the therapeutic population of Tregs is frozen in a cryovial in a volume of about 1.0 mL, about 1.1 mL, about 1.2 mL, about 1.3 mL, about 1.4 mL, about 1.5 mL, about 1.6 mL, about 1.7 mL, about 1.8 mL, about 1.8 mL, about 1.9 mL or about 2.0 mL.
- the therapeutic population of Tregs is frozen in a cryopreservation bag, e.g., a gas permeable bag.
- the therapeutic population of Tregs is frozen in a cryopreservation bag in a volume of about 1-2 mL, about 2- 5 mL, about 5-10 mL, about 10-15 mL, or about 15-20 mL.
- the therapeutic population of Tregs is frozen in a cryopreservation bag in a volume of about 1 mL, about 2 mL, about 5 mL, about 10 mL or about 20 mL.
- cryopreservation comprises decreasing the temperature of the therapeutic population of Tregs in the following increments: l°C/min to 4°C, 25°C/min to -40°C, 10°C/min to -12°C, l°C/min to -40°C, and 10°C/min to -80°C to -90°C.
- the cryopreserved therapeutic population of Tregs may be thawed, for example, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 1 wek, about 1-2 weeks, about 2-4 weeks, about 1 month, about 1-2 months, about 2-3 months, about 3-6 months, about 6-9 months, about 9-12 months, about 12-15 months, about 15-18 months, about 18-24 months, about 1-2 years, about 2-3 years, about 3-4 year or about 4-5 years after cryopreservation.
- the cryopreserved therapeutic population of Tregs may be thawed, for example, about 1 week, 1 month, about 3 months, about 6 months, about 9 months, about 12 months or about 18 months after cryopreservation.
- the cryopreserved therapeutic population of Tregs is thawed using a method that results in at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or greater than 95% viability of the Tregs as determined, for example, by Trypan Blue exclusion.
- the cryopreserved therapeutic population of Tregs is thawed and diluted in a solution comprising 0.9% sodium chloride. In some embodiments, the cryopreserved therapeutic population of Tregs is thawed and diluted in a solution comprising 0.9% sodium chloride and about 5% human serum albumin. In some embodiments, the cryopreserved therapeutic population of Tregs is thawed and diluted in a solution wherein the resulting solution comprises 0.9% sodium chloride. In some embodiments, the cryopreserved therapeutic population of Tregs is thawed and diluted in a solution wherein the resulting solution comprises 0.9% sodium chloride and about 5% human serum albumin. In certain embodiments, the cryopreserved therapeutic population of Tregs is thawed and withou further expansion is placed into a solution as described herein.
- the cryopreserved therapeutic population of Tregs is thawed and diluted in 50mL of a solution comprising 0.9% sodium chloride and about 5% human serum albumin. In some embodiments, the cryopreserved therapeutic population of Tregs is thawed and diluted in a solution, wherein the resulting solution is a 50mL solution comprising 0.9% sodium chloride and about 5% human serum albumin. In certain embodiments, the cryopreserved therapeutic population of Tregs is thawed and withou further expansion is placed into a solution as described herein.
- one cryovial containing the cryopreserved therapeutic population of Tregs is thawed and placed in 50mL of a solution comprising 0.9% sodium chloride and about 5% human serum albumin.
- one cryovial containing the cryopreserved therapeutic population of Tregs is thawed and placed in solution, wherein the resulting solution is a 50mL solution comprising 0.9% sodium chloride and about 5% human serum albumin.
- the cryopreserved therapeutic population of Tregs is thawed and withou further expansion is placed into a solution as described herein.
- the cryopreserved therapeutic population of Tregs is thawed using an automated thawing system (e.g., a COOK regentec thawing system).
- the cryopreserved therapeutic populations of Tregs is rapidly thawed.
- the cryopreserved therapeutic population of Tregs is thawed at a controlled rate.
- An exemplary thawing protocol is described in Section 8.6 below.
- the cryopreserved therapeutic population of Tregs is thawed in a water bath.
- the cryopreserved therapeutic population of Tregs is thawed at room temperature.
- the therapeutic population of Tregs is administered to a subject within about 2-10 hours, within about 4-8 hours, or within about 5-7 hours of thawing. In some embodiments, the therapeutic population of Tregs is administered to a subject within about 30 minutes, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 hours of thawing. In specific embodiments, the therapeutic population of Tregs is administered to the subject within about 6 hours of thawing.
- the cryopreserved therapeutic population of Tregs is thawed and administered to the patient without further dilution. In some embodiments, the cryopreserved therapeutic population of Tregs is thawed and administered to the patient without further dilution or further expansion. In some embodiments, the cryopreserved therapeutic population of Tregs is thawed and administered to the patient without further dilution and in combination with normal saline. In some embodiments, the cryopreserved therapeutic population of Tregs is thawed and administered to the patient without further dilution or further expansion and in combination with normal saline. In some embodiments, the thawed cryopreserved therapeutic population of Tregs and normal salineare administered concurrently and intravenously.
- no further expansion of the therapeutic population of Tregs is required between thawing and administration to the subject. In some embodiments, no further expansion of the therapeutic population of Tregs is performed between thawing and administration to the subject. 6.2.1. Automation
- a method of producing a therapeutic population of Tregs can be carried out in a closed system.
- the methods in some embodiments are carried out in an automated or partially automated fashion.
- the method of producing a therapeutic population of Tregs described herein e.g., the method described in Section 6.1, 8.3 or 8.5 herein
- the method is carried out in a G-REX ® culture system.
- Fig. 34 shows an exemplary process of producing a therapeutic population of Tregs ins a bioreactor.
- any one or more of the steps of the method of producing a therapeutic population of Tregs can be carried out in a closed system or under GMP conditions.
- one or more or all of the steps is carried out using a system, device, or apparatus in an integrated or self- contained system, and/or in an automated or programmable fashion.
- the system or apparatus includes a computer and/or computer program in communication with the system or apparatus, which allows a user to program, control, assess the outcome of, and/or adjust various aspects of the steps
- the method of producing a therapeutic population of Tregs comprises a step of expanding the Tregs.
- the expansion step is automated.
- the Tregs are expanded for 6, 7, 8, 9, 10, 11, or 12 days.
- the Tregs are expanded for 8 days. In another embodiment, the Tregs are expanded for 13, 14, or 15 days. In another embodiment, the Tregs are expanded for 15 days.
- the method of producing a therapeutic population of Tregs comprises administering an expansion agent (such as IL-2, a CD3 -activating agent and/or a CD28-activating agent) every 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
- the expansion agent such as IL-2, a CD3 -activating agent and/or a CD28-activating agent
- the expansion agent is first administered within 24h of initiating the culture.
- the expansion agent (such as IL-2, a CD3 -activating agent and/or a CD28-activating agent) is administered daily.
- the method of producing a therapeutic population of Tregs comprises replacing the culture medium every 1, 2, 3, 4, 5, 6 or 7 days.
- the media may be changed when the level of certain metabolites (e.g., lactacte) reach a predetermined threshold. In some embodiments, the media changes based on the expansion rate of the therapeutic population of Tregs.
- the concentration of cells in the culture is determined on Day 8. In some embodiments, the concentration of cells in the culture is determined on Day 15. In some embodiments, the system remains closed throughout the expansion step.
- compositions comprising a therapeutic population of Tregs suitable for administration to a subject.
- a cryopreserved composition comprising a therapeutic population of Tregs having characteristics as described herein.
- a therapeutic populatin of Tregs for example, a cryopreserved therapeutic population of Tregs, may be produced by the methods described herein.
- pharmaceutical compositions comprising a cryopreserved therapeutic population of Tregs that has been thawed and is present in a formulation suitable for administration to a subject, for example a human subject.
- a therapeutic population of Tregs provided herein comprises about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater than 95% viable cells as determined by trypan blue exclusion. In some embodiments, a therapeutic population of Tregs provided herein comprises more than about 70% viable cells.
- a therapeutic population of Tregs provided herein comprises about 1 x 10 6 to about 2 x 10 6 , about 2 x 10 6 to about 3 x 10 6 , about 3 x 10 6 to about 4 x 10 6 , about 4 x 10 6 to about 5 x 10 6 , about 5 x 10 6 to about 6 x 10 6 , about 6 x 10 6 to about 7 x 10 6 , about 7 x 10 6 to about 8 x 10 6 , about 8 x 10 6 to about 9 x 10 6 , about 9 x 10 6 to about 1 x 10 7 , about 1 x 10 7 to about 2 x 10 7 , about 2 x 10 7 to about 3 x 10 7 , about 3 x 10 7 to about 4 x 10 7 , about 4 x 10 7 to about 5 x 10 7 , about 5 x 10 7 to about 6 x 10 7 , about 6 x 10 7 to about 7 x 10 7 , about 7 x 10 7 to about 8 , about 8 x 10 6
- a therapeutic population of Tregs comprises about 1 x 10 6 to about 2 x 10 6 , about 2 x 10 6 to about 3 x 10 6 , about 3 x 10 6 to about 4 x 10 6 , about 4 x 10 6 to about 5 x 10 6 , about 5 x 10 6 to about 6 x 10 6 , about 6 x 10 6 to about 7 x 10 6 , about 7 x 10 6 to about 8 x 10 6 , about 8 x 10 6 to about 9 x 10 6 , about 9 x 10 6 to about 1 x 10 7 , about 1 x 10 7 to about 2 x 10 7 , about 2 x 10 7 to about 3 x 10 7 , about 3 x 10 7 to about 4 x 10 7 , about 4 x 10 7 to about 5 x 10 7 , about 5 x 10 7 to about 6 x 10 7 , about 6 x 10 7 to about 7 x 10 7 , about 7 x 10 7 to about 7 to about
- a therapeutic population of Tregs comprises about 1 x 10 6 to about 2 x 10 6 , about 2 x 10 6 to about 3 x 10 6 , about 3 x 10 6 to about 4 x 10 6 , about 4 x 10 6 to about 5 x 10 6 , about 5 x 10 6 to about 6 x 10 6 , about 6 x 10 6 to about 7 x 10 6 , about 7 x 10 6 to about 8 x 10 6 , about 8 x 10 6 to about 9 x 10 6 , about 9 x 10 6 to about 1 x 10 7 , about 1 x 10 7 to about 2 x 10 7 , about 2 x 10 7 to about 3 x 10 7 , about 3 x 10 7 to about 4 x 10 7 , about 4 x 10 7 to about 5 x 10 7 , about 5 x 10 7 to about 6 x 10 7 , about 6 x 10 7 to about 7 x 10 7 , about 7 x 10 7 to about 7 to about
- a therapeutic population of Tregs provided herein comprises lxlO 6 CD4+CD25+cells (+/- 10%) per kg of body weight of the subject as determined by flow cytometry. In some embodiments, a therapeutic population of Tregs provided herein comprises about or more than about 70% CD4+CD25+ cells and contains lxlO 6 CD4+CD25+cells (+/- 10%) per kg of body weight of the subject as determined by flow cytometry. The subject may be the same subject as the donor of the biological sample from which the Tregs were enriched.
- a cryopreserved therapeutic population of Tregs provided herein may comprise an increased proportion of O ⁇ 4 + O ⁇ 25 M ⁇ 1 Tregs relative to the proportion of CD4 _ CD25 high Tregs in a corresponding baseline Treg cell population, as determined by flow cytometry.
- a cryopreserved therapeutic population of Tregs comprises an increased proportion of CD4 + CD25 high CD127 low Tregs relative to the proportion of CD4+CD25 high CD127 low Tregs in the baseline Treg cell population, as determined by flow cytometry.
- an therapeutic population of Tregs provided herein may comprise an increased proportion of O ⁇ 4 + O ⁇ 25 w ⁇ 1 Tregs relative to the proportion of O ⁇ 4 + O ⁇ 25 M ⁇ 1 Tregs in the baseline Treg cell population,, as determined by flow cytometry.
- an expanded therapeutic population of Tregs comprises an increased proportion of CD4 + CD25 high CD127 low Tregs relative to the proportion of CD4+CD25 high CD127 low Tregs in the baseline Treg cell population, as determined by flow cytometry.
- “high” and “low” denote the expression level of a marker (e.g., CD25 or CD 127) in a subpopulation of cells relative to the entire population.
- the cryopreserved therapeutic population of Tregs comprises CD25 + Tregs wherein the expression of CD25 in the Tregs is increased relative to the expression of CD25 in the Tregs in the baseline Treg cell population, as determined by flow cytometry.
- the expression of CD25 is increased by at least about 5-fold, at least about 10-fold, at least about 15-fold, at least about 20-fold, at least about 25-fold, at least about 30- fold, at least about 35-fold, at least about 40-fold, at least about 45-fold or at least about 50- fold as determined by flow cytometry.
- the cryopreserved population of Tregs comprises CD127 + Tregs wherein the expression of CD127 in the Tregs is not increased greater than 3-fold relative to the expression of CD127 in the Tregs in the baseline Treg cell population,, as determined by flow cytometry. In some embodiments, the expression of CD127 is not increased relative to the expression of CD127 in the Tregs in the Treg-enriched cell population, as determined by flow cytometry.
- the cryopreserved therapeutic population of Tregs comprises at least 70%, at least 80%, or at least 90% CD4 + CD25 high CD127 low Tregs in the baseline Treg cell population, as determined by flow cytometry. In some embodiments, the cryopreserved therapeutic population of Tregs comprises at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% CD4 + CD25 + cells, as determined by flow cytometry. In some embodiments, a composition comprising a therapeutic population of Tregs provided herein comprises less than 20% CD8+ cells and comprises lxlO 6 CD4+CD25+cells (+/- 10%) per kg of body weight of the subject as determined by flow cytometry.
- a composition comprising a therapeutic population of Tregs provided herein comprises less than 20% CD8+ cells, comprises about or more than about 70% CD4+CD25+ cells, and comprises lxlO 6 CD4+CD25+cells (+/- 10%) per kg of body weight of the subject as determined by flow cytometry.
- the granularity of the Tregs in the cryopreserved therapeutic population of Tregs is increased relative to the granularity of the Tregs in the Treg- enriched cell population, as determined by flow cytometry. In some embodiments, the granularity of the Tregs is increased by at least about 1.5-fold, at least about 2-fold, or at least about 2.5-fold.
- the size of the Tregs in the cryopreserved therapeutic population of Tregs is increased relative to the size of the Tregs in the baseline Treg cell population, as determined by flow cytometry. In some embodiments, the size of the Tregs is increased by at least about 1.2-fold, at least about 1.5-fold, or at least about 2-fold.
- the cryopreserved therapeutic population of Tregs comprises CTLA4+ Tregs wherein the proportion of CTLA4+ Tregs is increased relative to the proportion of CTLA4+ Tregs in the baseline Treg cell population.
- an expanded therapeutic population of Tregs comprises CTLA4+ Tregs wherein the proportion of CTLA4+ Tregs is increased relative to the proportion of CTLA4+ Tregs in the baseline Treg cell population.
- the cryopreserved therapeutic population of Tregs comprises at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% CTLA4+ Tregs, as determined by flow cytometry.
- the Tregs in the cryopreserved therapeutic population of Tregs comprise FoxP3 -expressing Tregs wherein the expression of FoxP3 is increased in the Tregs relative to expression of FoxP3 in the Tregs in the baseline Treg cell population prior to expansion.
- a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein expresses high levels of FoxP3, wherein the one or more regulatory element (e.g., a promoter or an enhancer) of the FOXP3 gene is demethylated.
- the Treg specific demethylated region (TSDR) within FOXP3 is demethylated.
- the expression of one or more gene products associated with FOXP3 demethylation is increased in the therapeutic population of Tregs or in the cryopreserved therapeutic population of Tregs after expansion.
- one or more gene products associated with FOXP3 methylation is decreased in the therapeutic population of Tregs or in the cryopreserved therapeutic population of Tregs after expansion.
- a cryopreserved population of Tregs expresses high levels of glucocorticoid-induced tumor necrosis factor receptor (GITR).
- GITR glucocorticoid-induced tumor necrosis factor receptor
- a cryopreserved population of Tregs comprises less than 20% CD8+ cells, determined by flow cytometry.
- the viability of the cryopreserved therapeutic population of Tregs is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%, as determined by trypan blue staining performed following thawing of the cryopreserved therapeutic population. In some embodiments, the viability of the cryopreserved therapeutic population of Tregs, as determined by trypan blue staining performed following thawing of the cryopreserved therapeutic population, is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the viability of the expanded Treg cell population prior to the expanded Treg cell population being cryopreserved. In some embodiments, the viability of the therapeutic population of Tregs, as determined by trypan blue staining performed before cryopreserving the therapeutic population of Tregs is increased compared to the viability of the enriched Treg cell population prior to expansion.
- the suppressive function of the cryopreserved population of Tregs is at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%, as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation performed following thawing of the therapeutic population.
- Suppressive function of Tregs may be assessed, for example by measuring the proliferation of CFSE positive responder T cells by flow cytometry or thymidine incorporation.
- CFSE is an intracellular marker only present in the responder T cell population.
- Responder T cells are generally characterized as CD4 + CD25 T cells and may be isolated using the CD4+CD25+ Regulatory T Cell Isolation Kit (Miltenyi Biotec). For example, a sample maybe separated into a positively selected cell fraction containing CD4 + CD25 + regulatory T cells and the unlabeled CD4+CD25- cell effluent which contains the responder T cell population.
- the cryopreserved population of Tregs exhibits a suppressive function that is greater than the suppressive function of the enriched Treg cell population, as measured prior to expansion, as suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation.
- the suppressive function of the cryopreserved therapeutic population of Tregs is at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95% of the suppressive function of the expanded Treg cell population prior to the Tregs being cryopreserved, as determined by flow cytometry.
- the suppressive function of the therapeutic population of Tregs as determined by suppression of proliferation of responder T cells by flow cytometry or thymidine incorporation performed before cry opreserving the therapeutic population of Tregs is increased compared to the suppressive function of the enriched Treg cell population prior to expansion.
- the cryopreserved therapeutic population of Tregs exhibits an ability to suppress inflammatory cells, as measured by IL-6 production by the inflammatory cells (e.g., macrophages or monocytes from human donors or generated from induced pluripotent stem cells).
- the cryopreserved therapeutic population of Tregs exhibits an ability to suppress the function of myeloid cells (e.g., macrophages, monocytes, or microglia).
- the cryopreserved therapeutic population of Tregs exhibits an ability to suppress the release of inflammatory cytokines (e.g., IL-Ib, IL-6, IL-8, TNFa, or INGg).
- the expanded therapeutic population of Tregs exhibits an ability to suppress inflammatory cells, as measured by IL-6 production by the inflammatory cells (e.g., macrophages or monocytes from human donors or generated from induced pluripotent stem cells).
- the expanded therapeutic population of Tregs exhibits an ability to suppress the function of myeloid cells (e.g., macrophages, monocytes, or microglia).
- the expanded therapeutic population of Tregs exhibits an ability to suppress the release of inflammatory cytokines (e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy).
- the cryopreserved population of Tregs exhibits an improved ability to suppress secretion of inflammatory cytokines (e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy) from macrophages relative to the therapeutic population of Tregs prior to expansion.
- inflammatory cytokines e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy
- monocytes relative to the therapeutic population of Tregs prior to expansion.
- the cryopreserved population of Tregs exhibits an improved ability to suppress secretion of inflammatory cytokines (e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy) from microglia relative to the therapeutic population of Tregs prior to expansion.
- inflammatory cytokines e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy
- the expanded population of Tregs prior to cryopreservation exhibits an improved ability to suppress secretion of inflammatory cytokines (e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy) from macrophages relative to the therapeutic population of Tregs prior to expansion.
- inflammatory cytokines e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy
- monocytes relative to the therapeutic population of Tregs prior to expansion.
- the expanded population of Tregs prior to cryopreservation exhibits an improved ability to suppress secretion of inflammatory cytokines (e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy) from microglia relative to the therapeutic population of Tregs prior to expansion.
- inflammatory cytokines e.g., IL-Ib, IL-6, IL-8, TNFa, or INFy
- cryopreserved population of Tregs is thawed in accordance with a method described herein, e.g. a method described in Section 8.6 below.
- the therapeutic populations of Tregs and cryopreserved compositions comprising a therapeutic population of Tregs provided herein may be characterized by their gene product expression profiles.
- the gene product expression profile of a cryopreserved composition comprising a therapeutic population of Tregs provided herein may be compared to the Tregs at baseline.
- the term “baseline,” or “baseline Treg cell population denotes a population of Tregs that has been enriched from a patient sample but has not yet been expanded.
- the gene product expression profile is substantially the same in a cryopreserved composition comprising a therapeutic population of Tregs compared to the Tregs at baseline.
- two values are considered to be substantially the same when the difference between them is not statistically significant (i.e., p > 0.05) and/or if the binary log of the fold- difference between the two values is less than about 2-fold (increase or decrease).
- Changes in gene product expression may be expressed as “-fold increase” or “- fold decrease” or as the binary logarithm (or “log2”) of the -fold change.
- gene product expression is increased at least about 4-fold.
- gene product expression is increased about 5- 10-fold, about 10- 15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40- 45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90- fold, about 90- 100-fold, or at least about 100-fold.
- gene product expression is decreased at least about 4-fold.
- gene product expression is increased about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25- 30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60- fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold.
- the expression level of one or more gene product listed in Table 18 is detectable (i.e., above the level of detection for the method utilized such as single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- the expression level of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 gene products listed in Table 18 may be detectable (i.e., above the level of detection for the method utilized such as single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- the expression level of every gene product listed in Table 18 is detectable (i.e., above the level of detection for the method utilized such as single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- the expression level of one or more gene product listed in Table 18 is among the 50, 60, 70, 80, 90, 100, 110, 120, 130, 140 or 150 most highly- expressed gene products as determined by an unbiased proteomics analysis (e.g., single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- an unbiased proteomics analysis e.g., single-shot proteomics
- the expression level of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 gene products listed in Table 18 is among the 50, 60, 70, 80, 90, 100, 110, 120, 130, 140 or 150 most highly-expressed gene products as determined by an unbiased proteomics analysis (e.g., single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- an unbiased proteomics analysis e.g., single-shot proteomics
- the expression level of every gene product listed in Table 18 is among the 50, 60, 70, 80, 90, 100, 110, 120, 130, 140 or 150 most highly-expressed gene products as determined by an unbiased proteomics analysis (e.g., single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- an unbiased proteomics analysis e.g., single-shot proteomics
- the level of gene product expression may be above or below the limit of detection of the method being utilized to measure gene product expression.
- the expression level of a gene product listed in any of Table 12-Table 19 may be undetectable (i.e., below the level of detection for the method utilized such as single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- the expression level of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products listed in any of Table 12-Table 19 may be undetectable (i.e., below the level of detection for the method utilized such as
- the expression level of every gene product listed in any one of Table 12-Table 19 may be undetectable (i.e., below the level of detection for the method utilized such as single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- the expression level of a gene product listed in any of Table 12-Table 19 may be detectable (i.e., above the level of detection for the method utilized such as single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- the expression level of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products listed in any of Table 12-Table 19 may be detectable (i.e., above the level of detection for the method utilized such as single
- the expression level of every gene product listed in any one of Table 12-Table 19 may be detectable (i.e., above the level of detection for the method utilized such as single-shot proteomics) in a therapeutic population of Tregs or a cryopreserved composition comprising a therapeutic population of Tregs provided herein.
- the expression level of a gene product listed in any of Table 12-Table 19 may become detectable or undetectable in a therapeutic population of Tregs upon enrichment, expansion or cryopreservation.
- the expression level of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products listed in any of Table 12-Table 19 may become detectable or undetectable in a therapeutic population of Tregs upon enrichment, expansion
- Gene product expression may be determined by any method known in the art, for example, quantitative real-time PCR, FluidigmTM Chip assays, RNA sequencing, or proteomics analysis (e.g., single-shot proteomics).
- expression of one or more of the gene products listed in Table 12 and/or Table 13 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, or at least 90 of the gene products listed in Table 12 and Table 13 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of all the gene products listed in Table 12 and Table 13 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 12 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 gene products listed in Table 12 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 12 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 13 is decreased in the Tregs post-expansion compared to the Tregs at baseline. In some embodiments, expression of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 gene products listed in Table 13 is decreased in the Tregs post-expansion compared to the Tregs at baseline. In some embodiments, expression of every gene product listed in Table 13 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 12 and/or Table 13 that are not also listed in Table 19 is decreased in the Tregs post expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, or at least 90 of the gene products listed in Table 12 and Table 13 that are not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of all of the gene products listed in Table 12 and Table 13 that are not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 12 that are not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 gene products listed in Table 12 that are not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 12 that is not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 13 that are not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 of the gene products listed in Table 13 that are not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 13 that is not also listed in Table 19 is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 12 and/or Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80 at least 85, or at least 90 gene products listed in Table 12 and/or Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of every gene product listed in Table 12 and Table 13 that is not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 12 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 gene products listed in Table 12 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of every gene product listed in Table 12 that is not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 gene products listed in Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of every gene product listed in Table 13 that is not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 12 and/or Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, or at last 90 gene products listed in Table 12 and/or Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of every gene product listed in Table 12 and/or Table 13 that is not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of one or more of the gene products listed in Table 12 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, or at least 80 gene products listed in Table 12 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of every gene product listed in Table 12 that is not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of one or more of the gene products listed in Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 gene products listed in Table 13 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- the expression of every gene product listed in Table 13 that is not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs after expansion.
- expression of one or more of the gene products listed in Table 14-Table 18 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 14 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 15 is increased in the Tregs post-expansion compared to the Tregs at baseline. In some embodiments, expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 15 is increased in the Tregs post-expansion compared to the Tregs at baseline. In some embodiments, expression of every gene product listed in Table 15 is increased in the Tregs post-expansion compared to the Tregs at baseline. [00309] In some embodiments, expression of one or more of the gene products listed in Table 16 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, or at least 85 gene products listed in Table 16 is increased in the Tregs post-expansion compared to the Tregs at baseline. In some embodiments, expression of every gene product listed in Table 16 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 17 is increased in the Tregs post-expansion compared to the Tregs at baseline. In some embodiments, expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 17 is increased in the Tregs post-expansion compared to the Tregs at baseline. In some embodiments, expression of every gene product listed in Table 17 is increased in the Tregs post-expansion compared to the Tregs at baseline. [00311] In some embodiments, expression of one or more of the gene products listed in Table 18 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, or at least 35 gene products listed in Table 18 is increased in the Tregs post expansion compared to the Tregs at baseline. In some embodiments, expression of every gene product listed in Table 18 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 14-Table 18 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 14 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 14 that is not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 15 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 15 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 15 that is not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 16 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, or at least 85 gene products listed in Table 16 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 16 that is not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 17 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 17 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 17 that is not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of one or more of the gene products listed in Table 18 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, or at least 35 products listed in Table 18 that are not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- expression of every gene product listed in Table 18 that is not also listed in Table 19 is increased in the Tregs post-expansion compared to the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 14-Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products listed in Table 14- Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tre
- the expression of every gene product listed in Table 14-Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 14 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products listed in Table 14 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon
- the expression of every gene product listed in Table 14 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 15 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 15 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of every gene product listed in Table 15 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 16 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, or at least 85 gene products listed in Table 16 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of every gene product listed in Table 16 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 17 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 17 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of every gene product listed in Table 17 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, or at least 35 gene products listed in Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of every gene product listed in Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more of the gene products listed in Table 14-Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products listed in Table 14- Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tre
- the expression of every one of the gene products listed in Table 14-Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of one or more of the gene products listed in Table 14 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products listed in Table 14 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon
- the expression of every one of the gene products listed in Table 14 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of one or more of the gene products listed in Table 15 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 15 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of every one of the gene products listed in Table 15 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of one or more of the gene products listed in Table 16 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, or at least 85 gene products listed in Table 16 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of every one of the gene products listed in Table 16 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of one or more of the gene products listed in Table 17 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, or at least 30 gene products listed in Table 17 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of every one of the gene products listed in Table 17 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of one or more of the gene products listed in Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, or at least 35 gene products listed in Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- the expression of every one of the gene products listed in Table 18 that are not also listed in Table 19 is substantially the same in the cryopreserved therapeutic population of Tregs upon thawing as the expression of the one or more genes in the Tregs after expansion.
- one or more gene products listed in Table 12 and/or Table 13 is decreased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90-100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 12 and/or Table 13 is decreased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90-100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 12 is decreased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 12 is decreased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 13 is decreased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 13 is decreased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 14-Table 18 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20- 25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90-100- fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 14-Table 18 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50- 60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 14 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 14 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 15 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 15 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 16 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 16 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 17 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 17 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- one or more gene products listed in Table 18 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline.
- one or more gene products listed in Table 18 is increased at least about 4-fold, about 5-10-fold, about 10-15-fold, about 15-20-fold, about 20-25-fold, about 25-30-fold, about 30-35-fold, about 35-40-fold, about 40-45-fold, about 45-50-fold, about 50-60-fold, about 60-70 fold, about 70-80-fold, about 80-90-fold, about 90- 100-fold, or at least about 100-fold in cryopreserved Tregs relative to Tregs at baseline but the one or more gene products is substantially similar between cryopreserved Tregs and Tregs after expansion but before cryopreservation.
- the expression of one or more gene products associated with a dysfunctional Treg phenotype is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- a dysfunctional Treg phenotype includes, for example, dysregulated calcium dynamics, loss of MECP2 binding ability to 5-Hydroxymethylcytosine (5hmC)-DNA, dysregulation of MECP2 expression or activity, and loss of MECP2 regulation, phosphorylation or binding abilities.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products associated with a dysfunctional Treg phenotype (e.g., one or more of the gene products listed in Table 12) is
- the expression of one or more gene products associated with a dysfunctional Treg phenotype is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- a dysfunctional Treg phenotype includes, for example, dysregulated calcium dynamics, loss of MECP2 binding ability to 5hmC-DNA, dysregulation of MECP2 expression or activity, and loss of MECP2 regulation, phosphorylation or binding abilities.
- the expression of at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, at least 180, at least 190, at least 200, at least 210, at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290, at least 300, at least 310, at least 320, at least 330, at least 340, at least 350, at least 360, at least 370, at least 380, at least 390, or at least 400 gene products associated with a dysfunctional Treg phenotype (e.g., one or more of the gene products listed in Table 12) is
- the expression of one or more gene products associated with a dysfunctional Treg phenotype is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- a dysfunctional Treg phenotype includes, for example, dysregulated calcium dynamics, loss of MECP2 binding ability to 5hmC-DNA, dysregulation of MECP2 expression or activity, and loss of MECP2 regulation, phosphorylation or binding abilities.
- the expression of one or more gene products associated with a dysfunctional Treg phenotype is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- a dysfunctional Treg phenotype includes, for example, dysregulated calcium dynamics, loss of MECP2 binding ability to 5hmC-DNA, dysregulation of MECP2 expression or activity, and loss of MECP2 regulation, phosphorylation or binding abilities.
- the expression of one or more methylation- or epigenetics- associated gene products is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- the expression of one or more methylation- or epigenetics- associated gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more methylation- or epigenetics- associated gene products is decreased in the Tregs post-expansion compared to the Tregs at baseline.
- the expression of one or more methylation- or epigenetics- associated gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products known to be important for the proliferation, health, identification, and/or mechanism of Treg cells is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more gene products known to be important for the proliferation, health, identification, and/or mechanism of Treg cells is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products known to be important for the proliferation, health, identification, and/or mechanism of Treg cells is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more gene products known to be important for the proliferation, health, identification, and/or mechanism of Treg cells is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products known to be important for the proliferation, health, identification, and/or mechanism of Treg cells is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline and the expression of the same one or more gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products known to be important for the proliferation, health, identification, and/or mechanism of Treg cells is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline and the expression of the same one or more gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more mitochondria-r elated gene products is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more mitochondria-related gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more mitochondria-related gene products is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more mitochondria-related gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more mitochondria-related gene products is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline and the expression of the same one or more gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more mitochondria-related gene products is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline and the expression of the same one or more gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products associated with the cell cycle is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more gene products associated with the cell cycle is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products associated with the cell cycle is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline.
- the expression of one or more gene products associated with the cell cycle is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products associated with the cell cycle is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline and the expression of the same one or more gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- the expression of one or more gene products associated with the cell cycle is increased in a cryopreserved composition of Tregs relative to the expression of the one or more gene products in the Tregs at baseline and the expression of the same one or more gene products is substantially the same in the cryopreserved composition of Tregs relative to the expanded population of Tregs prior to cryopreservation.
- a composition provided herein is a pharmaceutical composition comprising a therapeutic population of Tregs and a carrier, excipient, or diluent. In some embodiments, a composition provided herein is a pharmaceutical composition comprising an effective amount of a therapeutic population of Tregs and a carrier, excipient, or diluent.
- the carrier, excipient, or diluent may be any pharmaceutically acceptable carrier, excipient or diluent, known in the art.
- pharmaceutically acceptable carriers include non-toxic solids, semisolids, or liquid fillers, diluents, encapsulating materials, formulation auxiliaries or carriers.
- a composition comprising a therapeutic population of Tregs provided herein further comprises a pharmaceutically acceptable carrier.
- a pharmaceutically acceptable carrier can include all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
- the pharmaceutical composition comprises a therapeutic population of Tregs suspended in a sterile buffer.
- a pharmaceutical composition provided herein comprises no more than 0.1% v/v, no more than 0.2% v/v, no more than 0.3% v/v, no more than 0.4% v/v, no more than 0.5% v/v, no more than 0.6% v/v, no more than 0.7% v/v, no more than 0.8% v/v, no more than 0.9% v/v, no more than 1% v/v, no more than 1.1% v/v, no more than 1.2% v/v, no more than 1.3% v/v, no more than 1.4% v/v or no more than 1.5% v/v DMSO.
- a composition comprising a therapeutic population of Tregs provided herein comprises no contaminants.
- a composition comprising a therapeutic population of Tregs provided herein comprises comprises a sufficiently low level of contaminants as to be suitable for administration, e.g., therapeutic administration, to a subject, for example a human subject.
- Contaminants include, for example, bacteria, fungus, mycoplasma, endotoxins or residual beads from the expansion culture.
- a composition comprising a therapeutic population of Tregs provided herein comprises less than about 5 EU/kg endotoxins.
- a composition comprising a therapeutic population of Tregs provided herein comprises about or less than about 100 beads per 3 x 10 6 cells.
- a composition comprising a therapeutic population of Tregs provided herein is sterile.
- isolation or enrichment of the cells is carried out in a closed or sterile environment, for example, to minimize error, user handling and/or contamination.
- sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
- kits for treatment comprising administering an effective amount of an expanded population of Tregs as described herein to a subject in need thereof.
- methods of treatment comprising administering an effective amount of an ex v/vo-expanded population of Tregs as described herein, e.g., as produced by the methods presented herein, to a subject in need thereof.
- kits for treatment comprising administering an effective amount of an expanded population of Tregs as described herein, wherein the population has been cryopreserved, to a subject in need thereof.
- methods of treatment comprising administering an effective amount of an ex v/vo-expanded population of Tregs as described herein, e.g., as produced by the methods presented herein, wherein the population has been cryopreserved, to a subject in need thereof.
- kits for treatment comprising administering an effective amount of an expanded population of Tregs as described herein, wherein the population has been cryopreserved, to a subject in need thereof, wherein the cryopreserved population is thawed and administered to the subject without further expansion.
- methods of treatment comprising administering an effective amount of an ex vivo- expanded population of Tregs as described herein, e.g., as produced by the methods presented herein, wherein the population has been cryopreserved, to a subject in need thereof, wherein the cryopreserved population is thawed and administered to the subject without further expansion.
- kits for treatment comprising administering an effective amount of a cryopreserved composition comprising a therapeutic population of Tregs to a subject in need thereof.
- methods for treating neurodegenerative disorders in a subject in need thereof comprising administering an effective amount of a cryopreserved composition comprising a therapeutic population of Tregs to a subject in need thereof.
- the cryopreserved population is thawed and administered to the subject without further expansion.
- the subject is diagnosed with or is suspected of having a disorder associated with Treg dysfunction. In some embodiments, the subject is diagnosed with or is suspected of having a disorder associated with Treg deficiency. In some embodiments, the subject is diagnosed with or is suspected of having a condition driven by a T cell response.
- the subject is diagnosed with or is suspected of having a neurodegenerative disease.
- the subject is diagnosed with or is suspected of having Alzheimer’s disease, Amyotrophic Lateral Sclerosis, Huntington’s disease, Parkinson’s disease, or frontotemporal dementia.
- the subject is diagnosed with or is suspected of having a disorder that would benefit from downregulation of the immune system.
- the subject is diagnosed with or suspected of having an autoimmune disease.
- the autoimmune disease may be, for example, systemic sclerosis (scleroderma), polymyositis, ulcerative colitis, inflammatory bowel disease, Crohn’s disease, celiac disease, multiple sclerosis (MS), rheumatoid arthritis (RA), Type I diabetes, psoriasis, dermatomyositis, systemic lupus erythematosus, cutaneous lupus, myasthenia gravis, autoimmune nephropathy, autoimmune hemolytic anemia, autoimmune cytopenia autoimmune hepatitis, autoimmune uveitis, alopecia, thyroiditis or pemphigus.
- the subject is diagnosed with or suspected of having heart failure or ischemic cardiomyopathy.
- the subject is diagnosed with or suspected of having graft-versus-host disease, e.g., after undergoing organ transplantation (such as a kidney transplantation or a liver transplantation), or after undergoing stem cell transplantation (such as hematopoietic stem cell transplantation).
- the subject is diagnosed with or suspected of having neuroinflammation.
- Neuroinflammation may be associated, for example, with stroke, acute disseminated encephalomyelitis (ADEM), acute optic neuritis, transverse myelitis, neuromyelitis optica (NMO), epilepsy, traumatic brain injury, spinal cord injury, encephalitis central nervous system (CNS) vasculitis, neurosarcoidosis, autoimmune or post-infectious encephalitis, or chronic meningitis.
- ADAM acute disseminated encephalomyelitis
- NMO neuromyelitis optica
- epilepsy traumatic brain injury, spinal cord injury, encephalitis central nervous system (CNS) vasculitis, neurosarcoidosis, autoimmune or post-infectious encephalitis, or chronic meningitis.
- the subject is diagnosed with or suspected of having cardo- inflammation, e.g., cardio-inflammation associated with atheroscleorosis, myocardial infarction, ischemic cardiomyopathy, with heart failure.
- cardo- inflammation e.g., cardio-inflammation associated with atheroscleorosis, myocardial infarction, ischemic cardiomyopathy, with heart failure.
- the subject is diagnosed with or suspected of having chronic inflammatory demyelinating polyradiculoneuropathy (CIDP).
- CIDP chronic inflammatory demyelinating polyradiculoneuropathy
- AIDP acute inflammatory demyelinating polyneuropathy
- the subject is diagnosed with or suspected of having Guillain-Barre syndrome (GBS).
- the subject has had a stroke.
- the subject is diagnosed with or suspected of having cancer, e.g., a blood cancer.
- cancer e.g., a blood cancer.
- the subject is diagnosed with or suspected of having asthma.
- the subject is diagnosed with or suspected of having eczema.
- the subject is diagnosed with or suspected of having a disorder associated with overactivation of the immune system.
- the subject is diagnosed with or suspected of having Tregopathy.
- the Tregopathy may be caused by a FOXP3, CD25, cytotoxic T lymphocyte- associated antigen 4 (CTLA4), LPS-responsive and beige-like anchor protein (LRBA), or BTB domain and CNC homolog 2 (BACH2) gene loss-of-function mutation, or a signal transducer and activator of transcription 3 (STAT3) gain-of-function mutation.
- CTL4 cytotoxic T lymphocyte- associated antigen 4
- LRBA LPS-responsive and beige-like anchor protein
- BACH2 BTB domain and CNC homolog 2
- STAT3 signal transducer and activator of transcription 3
- a cryopreserved composition comprising a therapeutic population of Tregs is administered within about 30 minutes, about lh, about 2-3h, about 3- 4h, about 4-5h, about 5-6, about 6-7h, about 7-8h, about 8-9h, or about 9-1 Oh of thawing the cryopreserved composition comprising a therapeutic population of Tregs.
- the cryopreserved composition comprising a therapeutic population of Tregs may be stored at about 2°C to about 8 °C (e.g., at about 4°) between thawing and administration.
- one dose of a therapeutic population of Tregs or a composition comprising a therapeutic population of Tregs is administered to a subject.
- a therapeutic population of Tregs or a composition comprising a therapeutic population of Tregs is administered more than once.
- a therapeutic population of Tregs or a composition comprising a therapeutic population of Tregs is administered two or more times.
- a therapeutic population of Tregs or a composition comprising a therapeutic population of Tregs is administered every 1- 2 weeks, 2-3 weeks, 3-4 weeks, 4-5 weeks, 5-6 weeks, 6-7 weeks, 7-8 weeks, 8-9 weeks, 9- 10 weeks, 10-11 weeks, 11-12 weeks, every 1-2 months, 2-3 months, 3-4 months, 4-5 months, 5-6 months, 6-7 months, 7-8 months, 8-9 months, 9-10 months, 10-11 months, 11-12 months, 13-14 months, 14-15 months, 15-16 months, 16-17 months, 17-18 months, 18-19 months, 19-20 months, 20-21 months, 21-22 months, 22-23 months, 23-24 months, every 1-2 years, 2-3 years, 3-4 years or 4-5 years.
- about lxlO 6 Tregs per kg of body weight of the subject are administered in the first administration and the number of Tregs administered is increased in the second third and subsequent administration. In some embodiments, about lxlO 6 Tregs per kg of body weight of the subject are administered in the first two administrations, and the number of Tregs administered is increased in every other administration thereafter (e.g., the 4 th , 6 th , 8 th and 10 th administration).
- about lxlO 6 Tregs per kg of body weight of the subject may be administered per month for the first and second month
- about 2xl0 6 Tregs per kg of body weight of the subject may be administered per month for the third and fourth month
- about 3xl0 6 cells per kg of body weight of the subject are administered per month for the fifth and sixth month.
- a method of treatment comprises administering a therapeutic population of autologous Tregs or a composition comprising a therapeutic population of autologous Tregs to the subject.
- a method of treating a neurodegenerative disorder in a subject comprises administering a therapeutic population of allogeneic Tregs or a composition comprising a therapeutic population of allogeneic Tregs to the subject.
- a subject treated in accordance with the method of treatment described herein further received one or more additional therapy or additional therapies.
- the subject is additionally administered IL-2.
- the dose of IL-2 may be about 0.5-lxl0 5 IU/m 2 , about l-1.5xl0 5 IU/m 2 , about 1.5-2xl0 5 IU/m 2 , about 2- 2.5xl0 5 IU/m 2 , about 2.5-3xl0 5 IU/m 2 , about 3 -3.5x10 5 IU/m 2 , about 3.5-4x10 5 IU/m 2 , about 4- 4.5xl0 5 IU/m 2 , about 4.5-5xl0 5 IU/m 2 , about 5-6xl0 5 IU/m 2 , about 6-7xl0 5 IU/m 2 , about 7- 8xl0 5 IU/m 2 , about 8-9xl0 5 IU/m 2 , about 9-10xl0 5 IU/m 2 , about 10-15xl0 5 IU/m 2 , about 15- 20xl0 5 IU/m 2 , about 15-
- the IL-2 is administered subcutaneously.
- the IL-2 may be administered at least 2 weeks, at least 3 weeks, or at least 4 weeks prior to the first Treg infusion.
- the subjected treated in accordance with the methods described herein receives one or more additional therapies are for the treatment of Alzheimer’s.
- Addition therapies for the treatment of Alzheimer’s may include acetylcholinesterase inhibitors (e.g., donepezil (Aricept @) , galantamine (Razadyne ® ), or rivastigmine (Exelon ® )) or NMD A receptor antagonists (e.g., Memantine (Akatinol ® , Axura ® , Ebixa ® /Abixa ® , Memox ® and Namenda ® ).
- Additional therapies may also include anti inflammatory agents (e.g., nonsteroidal anti-inflammatory drugs (NS AID) such as ibuprofen, indomethacin, and sulindac sulfide)), neuronal death associated protein kinase (DAPK) inhibitors such as derivatives of 3-amino pyridazine, Cyclooxygenases (COX-1 and -2) inhibitors, or antioxidants such as vitamins C and E.
- NS AID nonsteroidal anti-inflammatory drugs
- DAPK neuronal death associated protein kinase
- COX-1 and -2 Cyclooxygenases
- antioxidants such as vitamins C and E.
- a subject treated in accordance with the methods described herein receives on or more additional therapies for the treatment of ALS.
- Additional therapies for the treatment of ALS may include Riluzole (Rilutek ® ) or Riluzole (Rilutek ® )
- the therapeutic population of Tregs or the composition comprising a therapeutic composition of Tregs is administered to a subject by intravenous infusion.
- a method of treatment provided herein results in an increase in the Treg suppressive function in the blood from baseline. In some embodiments, a method of treatment provided herein results in an increase in the Treg suppressive function in the blood from baseline to week 4, week 8, week 16, week 24, week 30 or week 36. In some embodiments, a method of treatment provided herein results in an increase in the Treg suppressive function in the blood from baseline to week 24. In some embodiments, a method of treatment provided herein results in an increase in the Treg numbers in the blood from baseline. In some embodiments, a method of treatment provided herein results in an increase in the Treg numbers in the blood from baseline to week 4, week 8, week 16, week 24, week 30 or week 36. In some embodiments, a method of treatment provided herein results in an increase in the Treg numbers in the blood from baseline to week 24. [00401] The effect of a method of treatment provided herein may be assessed by monitoring clinical signs and symptoms of the disease to be treated.
- method of treatment provided herein results in a change in the Appel ALS score compared to baseline.
- the Appel ALS score measures overall progression of disability or altered function.
- the Appel ALS score decreases in a subject treated in accordance with a method provided herein compared to baseline, indicating an improvement of symptoms.
- the Appel ALS score remains unchanged ins a subject treated in accordance with a method provided herein compared to baseline.
- a method of treatment provided herein results in a change in the Amyotrophic Lateral Sclerosis Functional Rating Scale-revised (ALSFRS-R) score compared to baseline.
- the ALSFRS-R score assesses the progression of disability or altered function.
- the ALSFRS-R score increases in a subject treated in accordance with a method provided herein compared to baseline, indicating an improvement of symptoms.
- the Appel ALSFRS-R score remains unchanged in a subject treated in accordance with a method provided herein compared to baseline.
- a method of treatment provided herein results in a change in forced vital capacity (FVC; strength of muscles used with expiration) compared to baseline, where the highest number is the strongest measurement.
- FVC increases in a subject treated in accordance with a method provided herein compared to baseline.
- FVC remains unchanged in a subject treated in accordance with a method provided herein compared to baseline.
- a method of treatment provided herein results in a change in Maximum Inspiratory Pressure (MIP; strength of muscles used with inspiration) compared where the highest number is the strongest measurement.
- MIP increases in a subject treated in accordance with a method provided herein compared to baseline.
- MIP remains unchanged in a subject treated in accordance with a method provided herein compared to baseline.
- a method of treatment provided herein results in a change in Neuropsychiatric Inventory Questionnaire (NPI-Q) compared to baseline.
- NPI-Q provides symptom Severity and Distress ratings for each symptom reported, and total Severity and Distress scores reflecting the sum of individual domain scores.
- the NPI-Q score decreases in a subject treated in accordance with a method provided herein compared to baseline.
- NPI-Q score remains unchanged in a subject treated in accordance with a method provided herein compared to baseline.
- a method of treatment provided herein results in a decrease in the frequency of GI symptoms, anaphylaxis or seizures compared to baseline.
- a method of treatment provided herein results in a change in a change in CSF amyloid and/or CSF tau protein (CSF-tau) compared to baseline.
- CSF-tau CSF amyloid and/or CSF tau protein
- the levels of CSF amyloid and/or CSF tau protein decreases in a subject treated in accordance with a method provided herein compared to baseline.
- the levels of CSF amyloid and/or CSF tau protein remains unchanged in a subject treated in accordance with a method provided herein compared to baseline.
- a method of treatment provided herein results in a change in Clinical Dementia Rating (CDR) compared to baseline.
- CDR Clinical Dementia Rating
- the CDR rates memory, orientation, judgment and problem-solving, community affairs, home and hobbies, and personal care, and a global rating is then generated, ranging from 0-no impairment to 3- severe impairment.
- the CDR decreases in a subject treated in accordance with the methods provided herein compared to baseline.
- the CDR remains unchanged in a subject treated in accordance with a method provided herein compared to baseline.
- a method of treatment provided herein results in a change in Alzheimer's Disease Assessment Scale (ADAS)-cogl3 score compared to baseline.
- ADAS-cog tests cognitive performance and has an upper limit is 85 (poor performance) and lower limit is zero (best performance).
- the ADAS-cogl3 score decreases in a subject treated in accordance with a method provided herein compared to baseline.
- the ADAS-cog 13 score remains unchanged in a subject treated in accordance with a method provided herein.
- the efficacy of a method of treatment described herein may be assessed at about 20 weeks, about 24 weeks, about 28 weeks, about 32 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, about 56 weeks, about 60 weeks, about 64 weeks, about 68 weeks, about 72 weeks, about 76 weeks, about 80 weeks, about 84 weeks, about 88 weeks, about 92 weeks, about 96 weeks, about 100 weeks, at about 2-3 months, 3-4 months, 4-5 months, 5-6 months, 6-7 months, 7-8 months, 8-9 months, about 9-10 months, about 10-11 months, about 11-12 months, about 12-18 months, about 18-24 months, about 1- 2 years, about 2-3 years, about 3-4 years, about 4-5 years, about 5-6 years, about 6-7 years, about 7-8 years, about 8-9 years, or about 9-10 years after initiation of treatment in accordance with the method described herein.
- kits comprising a therapeutic composition of Tregs or a composition comprising a therapeutic population of Tregs provided herein.
- a kit provided herein comprises instructions for use, additional reagents (e.g., sterilized water or saline solutions for dilution of the compositions), or components, such as tubes, containers or syringes for collection of biological samples, processing of biological samples, and/or reagents for quantitating the amount of one or more surface markers in a sample (e.g., detection reagents, such as antibodies).
- additional reagents e.g., sterilized water or saline solutions for dilution of the compositions
- components such as tubes, containers or syringes for collection of biological samples, processing of biological samples, and/or reagents for quantitating the amount of one or more surface markers in a sample (e.g., detection reagents, such as antibodies).
- kits contain one or more containers containing a therapeutic population of Tregs or a composition comprising a therapeutic population of Tregs for use in the methods provided herein.
- the one or more containers holding the composition may be a single-use vial or a multi-use vial.
- the article of manufacture or kit may further comprise a second container comprising a suitable diluent.
- the kit contains instruction for use (e.g., dilution and/or administration) of a therapeutic population of Tregs or a composition comprising a therapeutic population of Tregs provided herein.
- Modification and changes may be made in the structure of the nucleic acids, or to the vectors comprising them, as well as to mRNAs, polypeptides, or therapeutic agents encoded by them and still obtain functional systems that contain one or more therapeutic agents with desirable characteristics.
- the resulting encoded polypeptide sequence is altered by this mutation, or in other cases, the sequence of the polypeptide is unchanged by one or more mutations in the encoding polynucleotide.
- the amino acid changes may be achieved by changing one or more of the codons of the encoding DNA sequence, according to the codon table, below.
- certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein’s biological functional activity, certain amino acid sequence substitutions can be made in a protein sequence, and, of course, its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. It is thus contemplated by the inventors that various changes may be made in the peptide sequences of the disclosed compositions or corresponding DNA sequences which encode said peptides without appreciable loss of their biological utility or activity.
- the hydropathic index of amino acids may be considered.
- the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982, incorporate herein by reference). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
- Each amino acid has been assigned a hydropathic index based on its hydrophobicity and charge characteristics (Kyte and Doolittle, 1982).
- an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent protein.
- substitution of amino acids whose hydrophilicity values are within ⁇ 2 is preferred, those within ⁇ 1 are particularly preferred, and those within + 0.5 are even more particularly preferred.
- amino acid substitutions are generally therefore based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
- Exemplary substitutions that take one or more of the foregoing characteristics into consideration are well known to those of ordinary skill in the art, and include arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
- Tregs suppress the proliferation of responder T lymphocytes and the activation of microglia.
- the expression of the Treg master transcription factor FOXP3 is reduced in rapidly progressing patients, with subsequent impairment of Treg suppressive function.
- FOXP3 expression and Treg suppressive function correlate with the extent and rapidity of disease progression.
- IL-2 and rapamycin Treg suppressive function is restored.
- phase I results demonstrated the safety and potential benefit of expanded autologous Treg infusions, thus providing further impetus for clinical trials employing these compositions in ALS patients.
- the results of the phase 1 clinical trial testing the safety of Treg infusions into ALS patients are described further below.
- Tregs Three ALS patients, with no family history of ALS, were selected based on their differing sites of disease onset and rates of progression. Participants underwent leukapheresis, and Tregs were subsequently isolated and expanded ex vivo. Tregs (1 x 10 6 cells/kg) were administered intravenously at early stages (4 doses over 2 months) and later stages (4 doses over 4 months) of disease. Concomitant interleukin (IL)-2 (2xl0 5 IU/m 2 /injection) was administered subcutaneously 3 times weekly over the entire study period. Participants were closely monitored for adverse effects and changes in disease progression rates. Treg numbers and suppressive function were assayed during and following each round of Treg infusions.
- IL interleukin-2
- Tregs were safe and well-tolerated in all participants.
- Treg numbers and suppressive function increased after each infusion.
- Measures of maximal inspiratory pressure also stabilized, particularly in two participants, during Treg infusions.
- CD4 + CD25 + FOXP3 + regulatory T lymphocytes are a subpopulation of T lymphocytes that are immunosuppressive and maintain tolerance to self-antigens, with their dysfunction playing a pivotal role in the development of autoimmune disorders (Refs. 1-4).
- Tregs In ALS mice, infusions of Tregs slow disease progression and prolong survival, and Tregs suppress the proliferation of responder T lymphocytes and the activation of microglia (Refs 5,6).
- the expression of the Treg master transcription factor FOXP3 is reduced in rapidly progressing patients (Ref. 7), with subsequent impairment of Treg suppressive functions; FOXP3 expression and Treg suppressive functions correlate with the extent and rapidity of disease progression (Ref.
- Treg infusions were administered every 2 weeks at an early stage of the disease followed by 4 Treg infusions administered every 4 weeks at a later stage.
- Each Treg dose was lxlO 6 cells/kg.
- the Treg dose was empirically determined, but was selected within the range of what has been shown to be safe and tolerable in patients with type 1 diabetes (Ref. 4).
- IL-2 was administered subcutaneously 3 times weekly at a dose of 2xl0 5 IU/m 2 /injection beginning the day after the first Treg infusion and continued throughout the study period.
- Tregs were isolated and expanded ex vivo in the Good Manufacturing Practice-compliant facility at M.D. Anderson Cancer Center according to a previously described protocol (Refs. 8,9). Each Treg infusion was administered intravenously through a peripheral line and participants were closely monitored for any infusion-related adverse responses for 4 hours following the infusion.
- ALSFRS-R ALS Functional Rating Scale
- AALS Appel ALS rating scale
- MIP maximal inspiratory pressure
- Peripheral blood was drawn one month prior to the first Treg infusion, immediately before each infusion, the day after each infusion, every 2 weeks during each round of infusions, and monthly after each round.
- the percentage of CD4 + CD25 + FOXP3 + Tregs within the total CD4 + population was assessed by flow cytometry (Ref. 8).
- Treg suppressive function on the proliferation of autologous responder T lymphocytes was assessed by [ 3 H]-thymidine incorporation (Ref. 8).
- Participant #2 dropped out of the study on week 50 due to his progressive disease and was placed in hospice care. He expired on week 51 due to respiratory failure secondary to ALS. Participant #3 developed two suspected gastrointestinal infections and an upper respiratory infection between weeks 24 and 29. She reported mild dyspnea on exertion beginning on week 48.
- Treg infusions were safe and well-tolerated regardless of the burden of disease.
- Treg suppressive function correlated with changes in the AALS; the greater the improvement in Treg suppressive function, the slower the rate of clinical progression. This correlation supports the value of Treg suppressive function as a meaningful indicator of clinical status.
- Treg infusions did not adversely affect respiratory function and appeared to stabilize the decline in MIPs in the two participants who were not being treated with noninvasive ventilation.
- low dose IL-2 administered subcutaneously for 1 year in 5 patients with ALS was safe and tolerable, but did not appear to alter the clinical course or increase endogenous Treg numbers, likely related to impaired endogenous Treg responsiveness to IL-2 (unpublished results).
- subcutaneous injections of low dose IL-2 were administered to stabilize the infused expanded Tregs.
- IL-2 was not of critical value in the interim between each round when Treg percentage and suppressive function, and clinical status deteriorated.
- Treg infusions Increased clinical progression rates were observed between each round of infusions, but it was not clear whether the progression was related to the cessation of Treg infusions or would have occurred spontaneously. More pertinent is the observation that subsequent Treg infusions were beneficial at later stages of disease with increased disease burden and rate of progression. Circulating functional Tregs may slow disease progression by suppressing peripheral proinflammatory monocytes/macrophages and responder T lymphocytes, as well as entering the CNS and suppressing activated microglia. Defining peripheral and central actions of Tregs merits further investigation.
- the Treg dose used in the phase I study was 1 x 10 6 /kg/infusion resulting in an average individual dose of 70-100 million cells per infusion. (See section 8.1 below). Eight doses were administered to each patient and each dose required its own manufacturing campaign that involved a 25-day ex vivo expansion, which drastically increased the efforts and costs per patient. Prolonged expansion times also result in Treg products with reduced suppressive capabilities, which would not be optimal for patients, e.g., patients with ALS. A successful manufacturing process would produce large numbers of highly functional Tregs over a shorter expansion time.
- Tregs From one patient exposure to leukapheresis with one expansion campaign, sufficient numbers of Tregs should be produced to accommodate larger phase 11/111 studies that require higher doses of Tregs over longer periods of time.
- sufficient numbers of Tregs should be produced for an entire therapeutic regimen, for example, an entire therapeutic regimen for treatment of a neurodegenerative disease such as ALS or Alzheimer’s disease, an autoimmune disease such as Type 1 diabetes or rheumatoid arthritis, or graft versus host disease (GVHD) such as GVHD following a bone marrow transplantation.
- a neurodegenerative disease such as ALS or Alzheimer’s disease
- an autoimmune disease such as Type 1 diabetes or rheumatoid arthritis
- GVHD graft versus host disease
- Treg expansion has been optimized and a more controlled manufacturing platform has been developed that yields vast numbers of highly functional Tregs that are cryopreserved, e.g., cryopreserved under cGMP conditions (see Sections 8.3, 8.5, and 8.6 below).
- Cryopreserved Tregs can be sent to patient locations where they can be thawed and infused into patients at different doses and frequencies under controlled conditions. Exemplary standard operating procedures for the manufacturing process are provided below, see, e.g., Sections 8.3, 8.5, and 8.6.
- Tregs As the role of Tregs in Alzheimer’s disease patients is poorly understood, the peripheral blood population and function of Tregs was evaluated during the disease course. It was found that Tregs exhibit a failure in suppressive activity at the clinical Alzheimer dementia stage, which could shift the immune system response towards a pro-inflammatory state, both in the periphery and in the brain. Further, the potential of ex vivo expansion of Tregs to restore homeostasis was also evaluated, as described below. 8.2.1. Materials and Methods
- Multicolor flow cytometry was used to assess the immunophenotype of Tregs.
- Antibodies against the following molecules were provided by: CD3 BV650 (BD Biosciences), CD8 BV450 (BD Biosciences), CD4 APC-H7 (BD Biosciences), CD25 PerCPCy5.5 (BD Biosciences), CD73 eFluor 450 (eBioscienceTM), PD-1 BV 650 (Biolegend). Dead cells were stained by LIVE/DEAD® Fixable Blue Dead Cell Stain Kit (Life Technology).
- Mononuclear immune cells were isolated from peripheral blood of participants using Lymphoprep (Stemcell) density gradient centrifugation. Tregs and responders T cells (Tresps) were isolated using the CD4 + CD25 + Regulatory T Cell Isolation Kit (Miltenyi Biotec) according to the manufacturer’s instructions. To increase purity, the positively selected cell fraction containing the CD4 + CD25 + regulatory T cells was run twice through the MS column. The unlabeled CD4 + CD25 cell effluent was collected as the Tresp population.
- Isolated Tresps were placed in a 96-well plate at a density of 50,000 cells per well followed by co-culture of corresponding Tregs at a ratio of 1 : 1 and 2: 1 (Tresps:Tregs) in at least triplicates.
- a CD3/CD28 T cell stimulation reagent (Miltenyi Biotec) was added to the co-cultures. After 5 days of culture, cells were pulsed with tritiated thymidine (1 Ci/well; Amersham Life Sciences) for 18 hours. The cells were harvested, and thymidine uptake was measured using a gas-operated-plate reader (Packard Instruments).
- Percentage suppression 100 - [(counts per minute of proliferating Tresps in the presence of Tregs /counts per minute of proliferating Tresps in the absence of Tregs) c 100]
- the protocol for generation of mature monocyte cells from induced pluripotent stem cells consists of 5 sequential steps through which mature monocytes are differentiated from human pluripotent cells in a stepwise manner (Yanagimachi etal, 2013). Mature monocytes were polarized to pro-inflammatory macrophages through treatment with GM-CSF (50 ng/mL) for 7 days, lipopolysaccharide (0.1 ng/mL) and interferon-g (0.2 ng/mL) for one hour. These iPSC-derived macrophages robustly produced proinflammatory IL-6, TNFa and IL-IB cytokines.
- GM-CSF 50 ng/mL
- lipopolysaccharide 0.1 ng/mL
- interferon-g 0.2 ng/mL
- Tregs from patients and healthy controls were co-cultured with activated macrophages for short (4 hours) and longer (24 hour) time periods. Cultured media was collected to assess cytokine protein levels via ELISA. Cells were then collected from culture and messenger RNA was isolated for the examination of cytokine transcripts.
- IL-13 and CD25 blockade in the co-culture anti-human IL-13 Ab (MAB2131) and anti-human CD25 Ab (MAB223) were used from R&D Systems. A falcon insert system (Life Science) was utilized to block direct contact between Treg and macrophage populations in the co-culture.
- Bead-selected CD4 + CD25 hlgh T lymphocytes were suspended at a concentration of 1 x 10 6 cells/mL in media containing 100 nM of rapamycin (Miltenyi Biotec), 500 IU/mL IL-2 and DynabeadsTM Human Treg Expander (GibcoTM) at a 4:1 bead-to-cell ratio.
- Fresh media containing rapamycin and IL-2 were added to the cells every 2 to 3 days. After 10 days of culture, cells were harvested and washed. The number of expanded Tregs and their suppressive functions were assayed.
- the minimum sample size was calculated to achieve 80% power with 5% significance level to detect the differences of Treg characteristic and function between varying stages of Alzheimer disease. Investigators were blinded to the identity of the groups during outcome assessment. Comparisons were performed using paired or unpaired student’s t-test (for two groups) or oneway ANOVA (for more than two groups). Correlations were determined using the linear regression. Data were expressed as Mean ⁇ SEM and p values less than 0.05 were considered significant. 8.2.2. Results
- Tregs were evaluated for the first time in Alzheimer disease. Following ex vivo expansion, the immunoregulatory capacity of Tregs was substantially enhanced in both patients and healthy controls through a cell contact-mediated mechanism. These expanded Tregs expressed 20-fold higher levels of CD25 protein on their cell surfaces.
- FIG. 4A A gating strategy (FIG. 4A) was applied that allows the identification of the Tregs by an existing established phenotype (CD4 + FoxP3 + CD25 high ). The percentage of CD4 + FOXP3 + CD25 high Tregs did not differ among HC, MCI and Alzheimer groups (FIG. 4B).
- CD4 + CD25 hlgh Tregs were positively selected from whole blood based on CD25 expression. Tregs and Tresps were co-cultured with varying numbers of Tregs against the same number of Tresps. First, the effects of age and gender on the suppressive function of Tregs were evaluated. There was no difference in the suppressive function of Tregs among men and women. No correlation was found between Treg suppressive activity and the age of the subjects (FIG. 5A).
- Tregs from the same individuals were expanded ex vivo in the presence of IL-2, rapamycin and CD3/CD28 beads for 10 days.
- CD4 + CD25 hlgh Tregs were co-cultured with pro- inflammatory iPSC-derived macrophages (Ml) and the relative changes of IL-6, TNFa, and IL-1B transcripts (after 4 hrs) and protein levels in the supernatant (after 24 hrs) were assayed.
- Baseline Tregs of MCI or Alzheimer patients showed no suppression on Ml- derived IL-6 transcript or protein (FIG. 7A and FIG. 7B).
- the Tregs from these same subjects were expanded ex vivo for 10 days and then co-cultured with pro-inflammatory macrophages.
- a reduction of Ml-derived IL-6 transcript and protein expression levels were noted following co-culture with expanded Tregs in all groups (FIG. 7A and FIG. 7B).
- the co-culture of baseline Tregs with Ml did not attenuate TNFa transcript and protein levels.
- Expanded Tregs of Alzheimer disease, MCI and HC displayed an enhanced capacity to suppress Ml-derived TNFa transcript and protein, compared to corresponding baseline Tregs (FIG. 7C and FIG.
- Tregs displayed an enhanced capacity to suppress Ml- derived IL1B transcript in all three groups (FIG. 7E).
- the protein values were too low to be measured reliably.
- Transcript levels of immunosuppressive cytokines known to be released from Tregs including TGFP, IL-10, IL-4, and IL-13, CD25, and immunomodulatory markers that require cell-to cell proximity or contact (PD1, CTLA4, Granzyme (GZM) A&B, PDL1, PDL2, CD39 and CD73) were examined by quantitative PCR.
- PD1, CTLA4, Granzyme (GZM) A&B, PDL1, PDL2, CD39 and CD73 immunomodulatory markers that require cell-to cell proximity or contact
- IL-13 and CD25 transcripts were up-regulated in expanded Tregs (FIG. 8A).
- the expression levels of PD1, PDL1, PDL2, CTLA4, CD39, CD73 and Granzyme A&B in Tregs were comparable between HC, MCI and AD at baseline. While there were no changes in CD39, PDL1, PDL2 and CTLA4 transcripts following ex vivo expansion, up-regulation of PD1, GZMB and CD73 transcripts in expanded Tregs were observed in all groups. Granzyme A expression levels were down-regulated in expanded Tregs (FIG. 8B).
- Tregs Treatment using expanded Tregs is a promising treatment for a wide variety of disoders, for example, neurodegenerative disorders such as ALS and Alzheimer’s disease, as described in Examples 1 and 2 above.
- technical challenges still prevent the wide application of Treg therapy.
- the methods for producing ex v/vo-expanded Treg cell populations, included cryopreserved therapeutic Treg cell populations, described in the present application address these challenges.
- the following is a list of representative steps illustrating an embodiment of the improved Treg manufacturing protocol.
- the exemplary protocol allows for the cryopreservation of a therapeutic population of Tregs after expansion, thus providing large amounts of Tregs at short notice, without the need for further expansion before administration.
- a more detailed summary of a representative Treg manufacturing protocol is presented at Fig. 14.
- the improved Treg manufacturing method may be used to expand Tregs from ALS patients. It is to be understood that the Treg production methods presented herein may also be applied to expand Tregs from other starting materials, including from cell samples from subjects with other disorders, e.g., other neurodegenerative disorders, or from healthy donor subjects.
- IL-2 is administered every 2-3 days on Days 6, 8 and 11. Cells are not discarded during expansion and thus, the scale is much larger requiring larger volumes of media.
- Cryopreservation is performed at the end of the process, which could be anywhere from Day 15 to Day 25 depending on when the expansion process reaches the dose needed to treat the patient for 1 year per a clinical protocol.
- Cryopreservation is performed manually on a large scale immediately following cell harvesting and bead removal.
- CryoStor® (CS10) freezing media is added at 4°C, and cells are transferred to cryovials within 10 minutes. Each cryovial represents 1 dose that would be thawed and administered back to the patient per the clinical protocol.
- Figs. 13 A and 13B show the granularity and size, respectively, of expanded and freshly isolated Tregs. These data indicate that expanded Tregs are larger than freshly isolated ones and that expanded Tregs have more granularity than freshly isolated ones.
- Regulatory T Cells from an ALS patient’s leukapheresis product.
- This protocol may also be applied to isolation and expansion of leukapheresis products from non- ALS subjects, e.g., healthy subjects, for example, as part of allogeneic Treg treatments, or patients exhibiting a different disorder, for example a different neurodegenerative disorder.
- FIG. 14 A process flow diagram is shown in FIG. 14.
- Processing is preceded by decontaminating the biological safety cabinet (BSC) surface and equipment with Deconquat (Veliek Associates Inc.) followed by 70% ethanol and cleaning all work surfaces with 70% isopropyl alcohol (IP A).
- BSC biological safety cabinet
- IP A isopropyl alcohol
- Step 01 Patient Leukapheresis Product Receiving
- IMPORTANT NOTE Leukapheresis products must be processed within 24 hours.
- Sterility 14 days aerobic and anaerobic culture: inoculate 0.5 mL of the leukapheresis product in each of the long-term culture bottles.
- STEP 02 Volume Reduction of Leukapheresis Products using the GE Healthcare/Biosafe Sepax 2 RM with the PeriCell Protocol and CS490.1 kit
- PeriCell input volume is 100 mL - 840 mL.
- Cells Density of leukapheresis product should be ⁇ 44 x 10 6 cells/mL.
- the minimum and maximum input volume is 100 mL - 840 mL
- the minimum and maximum output volume is 30 mL - 600 mL
- Kit ID Use barcode reader to enter kit ID from barcode on Tyvek cover of kit being used.
- the NeatCell protocol used with the CS-900.2 kit, is designed to process 30-120 mL starting material.
- the final product volume is 45 mL fixed volume.
- the NeatCell protocol will only accept a maximum input volume of 120 mL but it will remove and process up to 130 mL of the starting material. For high cell density sample, adjust the density following step below.
- NeatCell Specification Max Volume: 120 mL. Max Cell Density : 160 x 10 6 Cells/mL 8.5.4.3 Assembling the GE Healthcare Biosafe CS-900.2 kit
- CliniMACS CD19 Reagent 7.5 mL is sufficient for labeling up to 5 x 10 9 CD 19-positive cells out of up to 40 x 10 9 (40,000 x 10 6 ) leukocyte cells.
- Optimal combined labeling requires a sample volume of 87.5 mL plus the entire volume of one vial of CliniMACS CD8 Reagent and one vial of CliniMACS CD19 Reagent. [00567] The following information is needed to prepare Cells and Microbeads solution mix: Obtain Post NeatCell cell counts information.
- Target Cell Density ⁇ 400 x 10 6 cells/mL
- instrument screen refers to output of 1200 mL of target cells
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Cell Biology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Biomedical Technology (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Mycology (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Organic Chemistry (AREA)
- Biotechnology (AREA)
- Neurosurgery (AREA)
- Genetics & Genomics (AREA)
- Neurology (AREA)
- Environmental Sciences (AREA)
- Hematology (AREA)
- Dentistry (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Psychiatry (AREA)
- Hospice & Palliative Care (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Developmental Biology & Embryology (AREA)
- Virology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicinal Preparation (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
Claims
Priority Applications (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP20830430.3A EP4069259A2 (en) | 2019-12-05 | 2020-12-04 | Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease |
US17/292,423 US20230047159A1 (en) | 2019-12-05 | 2020-12-04 | Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease |
AU2020397893A AU2020397893A1 (en) | 2019-12-05 | 2020-12-04 | Regulatory T cell (Treg) compositions and methods for treating neurodegenerative disease |
MX2022006674A MX2022006674A (en) | 2019-12-05 | 2020-12-04 | Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease. |
JP2022534147A JP2023504734A (en) | 2019-12-05 | 2020-12-04 | Regulatory T cell (Treg) compositions and methods for treating neurodegenerative diseases |
CA3159411A CA3159411A1 (en) | 2019-12-05 | 2020-12-04 | Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201962944346P | 2019-12-05 | 2019-12-05 | |
US62/944,346 | 2019-12-05 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2021113685A2 true WO2021113685A2 (en) | 2021-06-10 |
WO2021113685A3 WO2021113685A3 (en) | 2021-07-08 |
Family
ID=74104193
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2020/063378 WO2021113685A2 (en) | 2019-12-05 | 2020-12-04 | Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease |
Country Status (7)
Country | Link |
---|---|
US (1) | US20230047159A1 (en) |
EP (1) | EP4069259A2 (en) |
JP (1) | JP2023504734A (en) |
AU (1) | AU2020397893A1 (en) |
CA (1) | CA3159411A1 (en) |
MX (1) | MX2022006674A (en) |
WO (1) | WO2021113685A2 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022261074A1 (en) * | 2021-06-08 | 2022-12-15 | The Methodist Hospital | Methods for producing regulatory t cell (treg) populations, treg compositions and methods for treatment |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4554101A (en) | 1981-01-09 | 1985-11-19 | New York Blood Center, Inc. | Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity |
-
2020
- 2020-12-04 US US17/292,423 patent/US20230047159A1/en active Pending
- 2020-12-04 CA CA3159411A patent/CA3159411A1/en active Pending
- 2020-12-04 WO PCT/US2020/063378 patent/WO2021113685A2/en unknown
- 2020-12-04 EP EP20830430.3A patent/EP4069259A2/en active Pending
- 2020-12-04 AU AU2020397893A patent/AU2020397893A1/en active Pending
- 2020-12-04 MX MX2022006674A patent/MX2022006674A/en unknown
- 2020-12-04 JP JP2022534147A patent/JP2023504734A/en active Pending
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4554101A (en) | 1981-01-09 | 1985-11-19 | New York Blood Center, Inc. | Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity |
Non-Patent Citations (25)
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022261074A1 (en) * | 2021-06-08 | 2022-12-15 | The Methodist Hospital | Methods for producing regulatory t cell (treg) populations, treg compositions and methods for treatment |
Also Published As
Publication number | Publication date |
---|---|
MX2022006674A (en) | 2022-11-14 |
JP2023504734A (en) | 2023-02-06 |
AU2020397893A1 (en) | 2022-06-09 |
WO2021113685A3 (en) | 2021-07-08 |
EP4069259A2 (en) | 2022-10-12 |
US20230047159A1 (en) | 2023-02-16 |
AU2020397893A8 (en) | 2022-06-23 |
CA3159411A1 (en) | 2021-06-10 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Liu et al. | Sinomenine inhibits the progression of rheumatoid arthritis by regulating the secretion of inflammatory cytokines and monocyte/macrophage subsets | |
ES2579762T3 (en) | Cancer treatment by combining lympho - reducing agent with CLTs and cytokines | |
JP2019013234A (en) | Stem cell microparticles | |
US20160143939A1 (en) | Compositions, kits and methods for treatment of cardiovascular,immunological and inflammatory diseases | |
ES2675317T3 (en) | Expansion of alloantigen reactive regulatory T lymphocytes | |
KR20190085000A (en) | Immunosuppressive mesenchymal cells and methods for their formation | |
AU2016250570A1 (en) | Therapeutic pooled blood apoptotic cell preparations and uses thereof | |
JP2016513095A (en) | Stem cell microparticles and miRNA | |
JP2015529450A (en) | Stem cell microparticles | |
TWI572718B (en) | Immunosuppressive cells and making methods and composition thereof | |
JP2015513403A5 (en) | ||
US20130302293A1 (en) | Compositions, cells, kits and methods for autologous stem cell therapy | |
US20220008474A1 (en) | Engineered regulatory t cells | |
Chang et al. | Single-cell transcriptomic identified HIF1A as a target for attenuating acute rejection after heart transplantation | |
WO2021113685A2 (en) | Regulatory t cell (treg) compositions and methods for treating neurodegenerative disease | |
JP6731114B2 (en) | Methods and compositions for the treatment of melanoma | |
Ren et al. | Rapamycin antagonizes angiogenesis and lymphangiogenesis through myeloid-derived suppressor cells in corneal transplantation | |
Guru et al. | HSF-1 enhances cardioprotective potential of stem cells via exosome biogenesis and their miRNA cargo enrichment | |
US20240093150A1 (en) | Improved t-cells for cancer therapy using amino acid starvation pathways | |
US8992933B2 (en) | Methods for activating T cells and modulating an immune response | |
Chan | Investigating the Effects of Polarized Monocytes/Macrophages in Osteoarthritis | |
Albericio et al. | Comparative Proteomic Analysis of Nuclear and Cytoplasmic Compartments in Human Cardiac Progenitor Cells. Functional Evaluation of IL1A and IMP3 | |
Schlaweck et al. | The CCR4/CCL17 axis drives intestinal acute Graft versus Host disease after allogeneic bone marrow transplantation | |
CA3210656A1 (en) | Regulatory t cell (treg) extracellular vesicle compositions and methods | |
WO2024018442A1 (en) | Preferential depletion of tcf1+ progenitor t-cells in severe covid-19 as mediated by interleukin 12 (il-12) |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 20830430 Country of ref document: EP Kind code of ref document: A2 |
|
ENP | Entry into the national phase |
Ref document number: 3159411 Country of ref document: CA |
|
ENP | Entry into the national phase |
Ref document number: 2022534147 Country of ref document: JP Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 2020397893 Country of ref document: AU Date of ref document: 20201204 Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2020830430 Country of ref document: EP Effective date: 20220705 |